Skip to main content

Full text of "USPTO Patents Application 10595161"

See other formats


UK Patent Application ,»,GB ,,,,2405793 ™A 

(43) Date of A Publication 18.03.2005 



(21) 


Application No: 


0321384.0 


(51) 


INT CL 7 : 










A61K 31/519 . A61P 37/06 39/00 


(22) 


Date of Filing: 


12.09.2003 












(52) 


UK CL (Edition X ): 








A5B BHA B180 B42Y B423 B48Y B480 B50Y B502 B504 


(71) 


Applicants): 






B51Y B511 B513 B54Y B541 B542 B55Y B552 B553 




4 AZA Bioscience nv 






B56Y B565 B566 B57Y B576 B58Y B586 B60Y B606 




(Incorporated in Belgium) 






B65Y B650 B657 B67Y B670 




Kapuctjnenvoer 33, B-3000 Leuven, 










Belgium 




(56) 


Documents Cited: 










WO 2000/039129 A1 DE 019944767 A 


(72) 


Inventor(s): 






JP 060192100 A JP 2003238409 A 




Piet Herdewijn 






US 5902810 A 




Mark Waer 










Steven Cesar Alfons De Jonghe 




(58) 


Field of Search: 




Un Yuan 






INT CL 7 A61K,A61P 




Sefrioui El Hassane 






Other WPI. EPODOC, JAPIO, CAS-ONUNE 


(74) 


Agent and/or Address for Service: 










Bird Goen & Co 










155 Regents Park Road LONDON, 










NW1 8BB, United Kingdom 









(54) Abstract Title: Pteridine derivatives for treating TNF-alpha related disorders 



(57) This invention relates to the use of a group of pteridine derivatives, their pharmaceutical^ acceptable 
salts, N-oxides, solvates, dihydro- and tetrahydroderivatives and enantiomers, for the manufacture of a 
medicament for the prevention or treatment of TNF-cc related disorders. The pterin preferably has an 
amino, or acetamino in the 2 position; hydroxy, an alkoxy, dialkylamino, dibenzylamino, a N containing 
heterocycle or an adamantylamino group in the 4 position; a hydrogen, styryl, or optionally substituted 
phenyl in the 6 position; and hydrogen, methyl or methoxy in the 7-position. The condition K may be 
septic or endotoxic shock, toxic effects of radiotherapy, TNF-ct or chemotherapeutic agents, or cachexia. 



O 
CD 

ro 

o 
ui 

CO 
CO 



Original Printed on Recycled Paper 



7s 



Figure 1 



CI 

H J 

R 2 ^ S N NH2 



XR, 



nV no 

R 2 ^N NH 2 



XR, 



I! X 
R 7 '' NH2 



XR t 



XR1 

N'VS 

6 



XR, 



A.1..J 



R 2 ^ N N 



XR, 



Figure 2 



R^N^NM, R^N^NH, R 2 ^N^NH 2 

9 10 11 

o n 3 

HON J 

« 13 



?1 
X 



k. JU 



R 2 N N 



Figure 4 



J J 



NH 2 

4 



H 3 C 



N ^ 



S N NH 2 



NH3 



O 

J-JU 



R 2 V N 



NH 2 
I 



IT •* 
R 2 N X N 



O 

J II 
R/" VV^R, 



NH 2 
R 2 N N 



NH 2 

R^N^NHj 



NH2 

R 2 '^n' NHj 



X 

) I 1 
R 2 'W 



V 



X 



R 2 A<V^R« 



Figure 5 



o 

X 

H 



► HN 



O 

7l JC 

H 



CI 



C | /V N" NH 2 



1 



X 

X " N 



i 1 

N V NH2 
X' * 



?1 
X 



nV NOj 

0 I 



X 

.JL 



I T :i 

A M ^ u ^ 



N N - Rj 



2405793 

1 

PTERIDINE DERIVATIVES FOR THE TREATMENT OF SEPTIC SHOCK 
AND TNF-a-RELATED DISEASES. 

FIELD OF THE INVFNTION 
5 The present invention relates to a novel medical indication of pteridine 
derivatives for the treatment of side effects of various chemotherapeutic drugs 
and/or of irradiation in cancer therapy. The present invention also relates to 
the use of polysubstituted pteridines for the prevention and/or the treatment of 
pathologic and inflammatory conditions such as septic shock, as well as toxic 

10 side effects, disorders and diseases related to or resulting from the exposure 
of patients to abnormally high levels of tumor necrosis factor-alpha 
(hereinafter referred as TNF-a) in general, and particularly following the 
administration of TNF-a in cancer treatment in humans. This invention also 
relates to the use of polysubstituted pteridines for the prevention and/or the 

15 treatment of radiotherapy-induced or chemotherapy-induced disorders such 
as mucositis, secondary myelodysplastic syndromes and radiation-induced 
graft-versus-host disease, and for the prevention and/or the treatment of 
injuries in cancer patients such as, but not limited to, apoptosis, radiation 
necrosis and nephrotoxicity following the administration of certain 

20 chemotherapeutic drugs such as cisplatin in cancer treatment Additionally the 
invention relates to the treatment of cachexia 

BACKGROUND OF THE INVENTION 

Several 2,4-diaminopteridine derivatives, including methotrexate are 
25 known in the art (for instance, see U.S. Patent No. 2,512,572) as being useful as 

antineoplastic agents 

Nevertheless, there still is a need in the art for specific and highly 

therapeutically active compounds, such as, but not limited to, drugs for 

preventing or treating cell proliferative disorders, including cancer. In 
30 particular, there is a need in the art to provide anti-cancer drugs which are 

active in a minor dose or in minimizing the side effects of other known and 

efficient anti-cancer drugs or radio-active treatments. 



10 



Septic shock is a major cause of death in intensive care units (about 
150,000 estimated deaths annually in the United States of America, despite 
treatment with intravenous antibiotics and supportive care) for which very little 
effective treatment is available at present. Patients with severe sepsis often 
experience failures of various systems in the body, including the circulatory 
system, as well as kidney failure, bleeding and clotting. Upopolysaccharide 
(hereinafter referred as LPS) is the primary mediator of Gramm-negative 
sepsis, the most common form of sepsis, by inducing the production of a 
whole array of macrophage-derived cytokines (such as TNF-a; interleukins 
such as IL-1, IL-6, IL-12; interferon-gamma (hereinafter referred IFN-y), etc.). 
These cytokines may induce other cells (e.g. T cells, NK cells) to make 
cytokines as well (e.g. IFN^y). In addition, other macrophage products (e.g. 
nitric oxide, hereinafter referred as NO) may also play a role in the 
pathogenesis of toxic shock. These substances (e.g. NO) may be induced 
15 directly due to microbial interactions or indirectly through the action of 
proinflammatory cytokines. LPS binds to a serum protein known as LPB and 
the LPS-LPB complex thus formed is recognized by the CD14 toll-like receptor 
4 (hereinafter referred as Tlr 4) complex on mononuclear phagocytes. Tlr4 is a 
signal transducing unit, the activation of which results in the release of 
mediators such as TNF-a, IL-1a, IL-ip and IL-6. These cytokines are 
important for the pathogenesis of shock. Their administration produces the 
clinical symptoms of septic shock and their blockade partially protects against 
LPS-induced lethal shock. 

Current therapeutic strategies for the treatment of septic shock are 
25 directed against LPS (e.g. antibodies against LPS or LBP-34-23) or against 
the cytokines induced by LPS (e.g. TNF antibodies) or against the receptor for 
LPS (e.a. CD14) Unfortunately the initial clinical data of these approaches are 
very disappointing and illustrate the redundancy of receptors and mediators 
involved in the pathogenesis of toxic shock. For instance flagellin seems to be 
30 another foxin that plays a role in Gramm-negative Salmonella shock syndrome 
and that cannot be prevented or treated by therapeutic strategies directed 
specifically at LPS. 



20 



3 



Clinical trials in humans with TNF-a blocking antibodies (such as the IL- 
1 receptor antagonist or PAF receptor antagonists) have been unsuccessful 
yet, as have been approaches to down regulate inflammation (e.g. using 
prednisolone) or to block endotoxins. These products must be administered 
5 very early after the onset of the disease, which is in most cases not possible. 

The only drug currently approved by health authorities for the treatment 
of adult patients with the most serious forms of sepsis, including septic shock, 
is a genetically engineered version of a naturally occurring human protein, 
Activated Protein C, known as Xigris® or drotecogin-alpha which shows only 

10 moderate efficacy. Furthermore, because Activated Protein C interferes with 
blood dotting, the most serious side effect associated with Xigris® is bleeding, 
including bleeding that causes stroke Thus Xigris® is contra-indicated for 
patients who have active internal bleeding, or who are more likely to bleed 
because of certain medical conditions including recent strokes, recent head or 

15 spinal surgery or severe head trauma. Beacause treatment with Xigris® comes 
with potentially serious risks, the benefits and risks of treatment with Xigris® 
must be carefully weighed for each individual patient. 

Therefore there is a strong need in the art for new medications, either 
alone or in combination with the currently suggested treatments, for treating 

20 the most serious forms of life-threatening illnesses caused by severe infection, 
such as septic shock. 

TNF-a is generally considered to be the key mediator in the mammalian 
response to bacterial infection. It is a strong pro-inflammatory agent that will 
affect the function of almost any organ system, either directly or by inducing 

25 the formation of other cytokines like IL-1 or prostaglandines TNF-a is also a 
potent anti-tumor agent. If administered in small quantities to humans, it 
causes fever, headache, anorexia, myalgia, hypotension, capillary leak 
syndrome, increased rates of lipolysis and skeletal muscle protein degradation 
(including cachexia). Its use in cancer treatment is therefore very much limited 

30 by its severe side effects. 

TNF-a, a pleiotropic cytokine produced mainly by activated 
macrophages, exerts an in vitro cytotoxic action against transformed cells and 
in vivo anti-tumor activities in animal models. However, despite the fact that 



TNF-a is used in cancer patients especially to treat melanoma and sarcoma, 
the major problem hampering its use is toxicity. Indeed, TNF-a induces shock- 
like symptoms such as bowel swelling and damage, liver cell necrosis, 
enhanced release of inflammatory cytokines such as IL-1 or IL-6. and 
5 hypotension probably due to the release of inducers of vessels dilatation such 
nitric oxide and other proinflammatory cytokines. Cardiovascular toxicity is 
usually dose-limiting. Hypotension can be severe with systolic Wood pressure 
below 60 mm Hg. Respiratory compromise is common after treatment with 
TNF-a and may require mechanical ventilation. Upper as well as lower 
10 digestive tract symptoms are also common in this type of treatment. Nausea 
and vomiting can be distressing and in some cases dose-limiting ; Watery 
diarrhea is frequently observed. Neurological sequelae of treatment with TNF- 
a can also occur. 

Hence, compounds that inhibit the toxic effects of TNF-a but that do not 
1 5 inhibit TNF-a anti-tumor effect are highly desirable for the treatment of cancer 
patients. Presently, several clinical trials involving TNF-a are being developed 
for the cancer of organs such as liver, lung, kidney and pancreas, which are 
based on a procedure including the steps of organ isolation, injection of TNF- 
a into the isolated organ, and reperfusion of the treated organ. However, even 
20 for isolated organ perfusion, some TNF-a usually escapes to the general 
blood circulation and leads to the mortality of about 10% of the patients thus 
treated. Many patients treated by this procedure also require intensive care 
unit rescue to cope with the toxic side-effects of such TNF-a treatment. 

Combined treatment of TNF-a with alkylating drugs in an isolated organ 
25 perfusion model has received considerable attention. TNF-a is currently 
successfully used in isolated limb perfusion of human cancer patients and, in 
combination with melphalan and interferon-gamma, against melanoma, 
sarcomas and carcinomas. 

The gastrointestinal mucosa is very sensitive to chemotherapeutic 
30 drugs Mucositis caused by chemotherapy usually begins rapidly after 
initiation of the treatment with inflammation and ulceration of the 
gastrointestinal tract and leading to diarrhea. Severe, potentially life- 



threatening, diarrhea may require interruption of the chemotheraputic 
treatment and subsequent dose reduction of the therapeutic agent. The oral 
cavity is often the place of severe side effects from cancer therapy that 
adversely affects the quality of life of the patient and its ability to tolerate the 
5 therapy. These side effects can be caused by radiotherapy as well as 
chemotherapy. A relationship between both serum and mucosal levels of 
TNF-o and IL-1 correlates with nonhematologic toxicities, including mucositis. 

Radiation injuries occurring e.g. after a single high-dose irradiation 
include apoptosis as well as radiation necrosis. Even normal tissues protected 
by shielding during irradiation may be considerably damaged. It was found in 
experimental animal models that the radiation injuries after a single high-dose 
irradiation typically used for the treatment of various malignant tumors consist 
of radiation necrosis and apoptosis, which were correlated with the expression 
of TNF-a and TGF-pi . 

Irradiation may induce graft-versus-host disease (hereinafter referred 
as GVHD) in cancer patients. This disease may occur especially in patients 
receiving allogeneic* bone marrow transplantation as a treatment for cancers 
such as leukemia or lymphoma and can lead to the death of about 25% of the 
relevant patients. Before bone marrow transplantation, leukaemia patients for 
example receive either total body or total lymphoid irradiation to suppress their 
immune system. However, such irradiation induces not only necrosis but also 
the release of proinflammatory cytokines mainly TNF-a, IL-1 and IL-6 which in 
turn induce direct host tissues inflammation and activation of donor cells 
against host antigens leading to GVHD. 

Cisplatin is an effective chemotherapeutic agent used in the treatment 
of a wide variety of both pediatric and adult malignancies, including testicular, 
germ cell, head and neck (cervical), bladder and lung cancer. Dose- 
dependent and cumulative nephrotoxicity is the major side effect of cisplatin, 
sometimes requiring a reduction in dose or discontinuation of the treatment. 
Other side effects of cisplatin include kidney damage, loss of fertility, harmful 
effect on a developing baby, temporary drop in bone marrow function causing 
drop in white blood cell count, anaemia, drop in platelets causing bleeding, 



loss of appetite, numbness or tingling in limbs, loss of taste, allergic reactions, 
and hearing disorders (difficulty in hearing some high-pitched sounds, 
experiencing ringing in the ears). Blurred vision may also be a side effect with 
high doses of cisplatin. It was shown that TNF-a is a key element in a network 
5 of proinflammatory chemokines and cytokines activated in the kidney by 
cisplatin. Blockade of TNF-a action would prevent the activation of this 
cytokine network and would provide protection against cisplatin nephrotoxicity. 
Hence, compounds that inhibit the toxic effects of cisplatin but that do not 
inhibit cisplatin anti-tumor effects are highly desirable for the treatment of 
10 cancer patients. 

A surplus of TNF-a also causes a dramatic change of endothelial cells. 
In particular, TNF-a is an important mediator of skeletal muscle degeneration 
associated with cachexia, a debilitating syndrome characterized by extreme 
weight loss and whole-body wasting. Cachexia is usually a secondary 
15 condition whereby there is excessive tissue catabolism in combination with 
deficient anabolism. It is frequently seen in patients afflicted with chronic 
diseases such as cancer, cardiopulmonary diseases, aging, malabsortive 
disorders, excessive physical stress, easting disorders and acquired 
immmuno-deficiency syndrome (AIDS). Some authors consider that the 
20 elevated TNF-a values found in at least 50% of cancer patients in the active 
stage of the disease can result in cachexia. TNF-a levels in clinically healthy 
adults, as well as in adult cancer patients, are well documented, for instance 
by Nenova et al in Archives of Hellenic Medicine (2000) 17:619-621. Serum 
TNF-a concentrations in healthy children as well as in children with 
25 malignancies are documented for instance by Saarinen et al. in Cancer 
Research (1990) 50:592-595. A very significant proportion of cancer 
mortalities result from cachexia rather than from tumor burden. Chronic 
wasting disease (cachexia) may result when excessive cellular damage 
results in the release of substances (TNF-a, collagenase, hyaluronidase) that 
30 further catabolize the so-called healthy tissue resulting in an inability to 
assimilate nutrients required for anabolic restructuring of associated tissue. 

Infants infected with human immunodeficiency virus type 1 (HIV-1) 
show growth retardation and severe weight loss that can lead to death. The 



7 



overproduction of certain cytokines has been implicated as a possible cause 
for this. For instance, according to Rautonen et al. in AIDS (1991) 5:1319- 
1325, serum IL-6 concentrations are elevated and associated with elevated 
TNF-o concentrations in children with HIV infection Swapan et al. in Journal 
5 of Virology (2002) 76. 1 1 71 0-1 1 71 4 have shown that reduction of TNF-a levels 
by either anti-TNF-a antibodies or human chorionic gonadotropin inhibits the 
expression of HIV-1 proteins and prevents cachexia and death. 

Very few drugs have been suggest at present for the treatment of 
cachexia. Some high-dose progestins like megestrol acetate, an agent used 

10 for the treatment of metastatic breast cancer, and medroxyprogesterone 
acetate were shown in randomized clinical trials to provide a statistically 
significant advantage as regards improved appetite and body weight gain 
Hence, compounds that stimulate appetite and body weight gain without 
inhibiting the anti-tumor effect or anti-viral effect of co-administered drugs are 

IS highly desirable for the treatment of cachexia. More specifically, there is a 
need in the art for treating cachexia by the administration of compounds that 
reduce TNF-a levels in the serum of humans. 

TNF-a is also suspected to play a role, through a possible dual action in 
the hematopoietic environment, in the development of hematologic 

20 malignancies such as idiopathic myelodysplastic syndromes occurring most 
often in elderly people but also occasionally in children, these syndromes 
being currently regarded as the early phase of acute leukemia. 

There is a strong need in the art to improve, or to provide alternatives 
to, the existing prophylactic or therapeutic solutions to all the aforesaid 

25 diseases. Meeting this need in the art constitutes the main goal of the present 
invention. 



30 



SUMMARY OF THE INVENTION 

The present invention relates to the unexpected finding that a class of 
pteridine derivatives having the general formula (I): 



wherein X represents an oxygen atom or a group with the formula S(0)m 
wherein m is an integer from 0 to 2, or a group with the formula NZ and wherein: 
- Ri is a group selected from the group consisting of C1-7 alkyl, C2-7 
alkenyl, C 2 . 7 alkynyl, C^o cycloalkyl, C^o cycloalkenyl, aryl, alkylaryl, 
arylalkyl, heterocyclic, heterocyclic-substituted alkyl and alkyl- 
substituted heterocyclic, each of said groups being optionally 
substituted with one or more substituents selected from the group 
consisting of halogen, C1-4 alkyl, Cm alkoxy, C 2 -? alkenyl, C 2 . 7 alkynyl, 
halo Ci^j alkyl, C3-10 cycloalkoxy, aryloxy, arylalkyioxy, oxyheterocydic, 
heterocyclic-substituted alkyloxy, thio C1.7 alkyl, thio C^o cycloalkyl, 
thioaryl, thioheterocyclic, arylalkylthio, heterocyclic-substituted alkylthio, 
formyl, hydroxyl, suHhydryl, nitro, hydroxylamino, mercaptoamino, 
cyano, carboxylic aad or esters or thioesters or amides or thioamides 
or halides or anhydrides thereof, thiocarboxylic acid or esters or 
thioesters or amides or thioamides or halides or anhydrides thereof, 
carbamoyl, thiocarbamoyl, ureido, thio-ureido, amino, cydoalkylamino, 
alkenylamino, cycloalkenylamino, alkynylammo, arylamino, arylalkyl- 
amino, hydroxylalkylamino, mercaptoalkylamino, heterocyclic amino, 
hydrazino, alkylhydraano and phenylhydrazino; or Ri is a carboxyalkyl, 
carboxyaryl, thiocarboxyaryl or thiocarboxyalkyl group; 

- Z is a group independently defined as Ri or Z is hydrogen or the group 
NZ together with Rt is either hydroxylamino or an optionally substituted 
heterocyclic group containing at least one nitrogen atom; 

- R 2 is selected from the group consisting of amino; acyiamino; 
thioacylamino, carbamoyl; thiocarbamoyl, ureido; thioureido, sulfon- 
amide; hydroxylamino; alkoxyamino; thioalkylamino; mercaptoamino, 
hydrazino; alkylhydrazino; phenylhydrazino; optionally substituted 
heterocyclic radicals; Ci_ 7 alkylamino; arylamino; arylalkylamino; 
cydoalkylamino; alkenylamino; cydoalkenylamino; heterocydic amino; 



hydroxyalkylamino; mercaptoalkylamino; C1.7 alkoxy; C3-10 cycloalkoxy; 
thio C1.7 alkyl; arylsulfoxide; arylsulfone; heterocyclic sulfoxide; 
heterocyclic sulfone; thio C3-10 cycloalkyl; aryloxy; arylthio; arylalkyloxy; 
arylalkylthio; oxyheterocyclic and thioheterocyclic radicals, 
- R4 is an atom or a group selected from the group consisting of 
hydrogen; halogen; C1.7 alkyl; C 2 - 7 alkenyl; C 2 - 7 alkynyl; halo C1-7 alkyl; 
carboxy C,. 7 alkyl; acetoxy C1-7 alkyl; carboxyaryl; C,. 7 alkoxy; C3.10 
cycloalkoxy; aryloxy; arylalkyloxy; oxyheterocyclic; heterocyclic- 
substituted alkyloxy; thio C1.7 alkyl; thio C3-10 cycloalkyl; thioaryl; 
thioheterocyclic; arylalkylthio; heterocyclic-substituted alkylthio; amino; 
hydroxylamino; mercaptoamino, acylamino; thioacylamino; alkoxy- 
amino; thioalkylamino; acetal; thioacetal; carboxylic acid; carboxylic 
acid esters, thioesters, halides, anhydrides, amides and thioamides; 
thiocarboxylic aad; thiocarboxylic acid esters, thioesters, halides, 
anhydrides, amides and thioamides; hydroxyl, sulfhydryl; nitro; cyano; 
carbamoyl; thiocarbamoyl, ureido; thio-ureido; alkylamino; 
cycloalkylamino; alkenylamino; cycloalkenylamino; alkynylamino; 
arylamino; arylalkylamino; hydroxyalkylamino; mercaptoalkylamino; 
heterocyclic amino; heterocyclic-substituted alkylamino; oximino; 
alkyloximino; hydrazino; alkylhydrazino; phenylhydrazino; cysteinyl 
acid, esters, thioesters, halides, anhydrides, amides and thioamides 
thereof; aryl groups optionally substituted with one or more substituents 
selected from the group consisting of halogen, C1.7 alkyl, C1.7 alkoxy, 
C2-7 alkenyl, C2-7 alkynyl, halo C1.7 alkyl, nitro, hydroxyl, sulfhydryl, 
amino, C3-10 cycloalkoxy, aryloxy, arylalkyloxy, oxyhetero-cyclic, 
heterocyclic-substituted alkyloxy, thio C1.7 alkyl, thio C3-10 cycloalkyl, 
thioaryl, thioheterocyclic, arylalkylthio, heterocyclic-substituted alkylthio, 
formyl, carbamoyl, thiocarbamoyl, ureido, thio-ureido, sulfonamido, 
hydroxylamino, alkoxyamino, mercaptoamino, thioalkylamino, acyl- 
amino, thioacylamino, cyano, carboxylic acid or esters or thioesters or 
halides or anhydrides or amides thereof, thiocarboxylic acid or esters or 
thioesters or halides or anhydrides or amides thereof, alkylamino, 
cycloalkylamino, alkenylamino, cycloalkenylamino, alkynylamino, 



10 



arylamino, arylalkylamino, hydroxyalkylamino, mercaptoalkylamino, 
heterocyclic amino, hydrazino, alkyl-hydrazino and phenylhydrazino; 
optionally substituted heterocyclic radicals; aromatic or heterocyclic 
substituents substituted with an aliphatic spacer between the pteridine 
ring and the aromatic or heterocyclic substituent, whereby said aliphatic 
spacer is a branched or straight, saturated or unsaturated aliphatic 
chain of 1 to 4 carbon atoms which may contain one or more functions, 
atoms or radicals selected from the group consisting of carbonyl (oxo), 
thiocarbonyl, alcohol (hydroxyl), thiol, ether, thio-ether, acetal, thio- 
acetal, amino, imino, oximino, alkyloximino, amino-acid, cyano, 
acylamino, thioacylamino, carbamoyl, thiocarbamoyl, ureido, thio- 
ureido, carboxylic acid or ester or thioester or halide or anhydride or 
amide, thiocarboxylic acid or ester or thioester or halide or anhydride or 
amide, nitro, thio Ci_ 7 alkyl, thio C3-10 cycloalkyl, hydroxylamino, 
mercaptoamino, alkylamino, cycloalkylamino, alkenylamino, cydo- 
alkenylamino, alkynylamino, arylamino, arylalkylamino, hydroxyalkyl- 
amino, mercaptoalkylamino, heterocyclic amino, hydrazino, 
alkylhydrazino, phenylhydrazino, sulfonyl, sulfinyl, sulfonamido and 
halogen; branched or straight, saturated or unsaturated aliphatic chains 
of 1 to 7 carbon atoms optionally containing one or more functions 
selected from the group consisting of carbonyl (oxo), thiocarbonyl, 
alcohol (hydroxyl), thiol, ether, thioether, acetal, thio-acetal, amino, 
imino, oximino, alkyloximino, amino-acid, cyano, acylamino; 
thioacylamino; carbamoyl, thiocarbamoyl, ureido, thio-ureido, carboxylic 
acid ester or halide or anhydride or amide, thiocarboxylic acid or ester 
or thioester or halide or anhydride or amide, nitro, thio C1.7 alkyl, thio 
C3-10 cycloalkyl, hydroxylamino, mercaptoamino, alkylamino, cyclo- 
alkylamino, alkenylamino, cycloalkenylamino, alkynylamino, arylamino, 
arylalkylamino, hydroxyalkylamino, mercapto-alkylamino, heterocyclic 
amino, hydrazino, alkylhydrazino, phenyl-hydrazino, sulfonyl, sulfinyl, 
sulfonamido and halogen; and 



11 

- F*3 is an atom or a group defined as or R3 together with R4 forms a 
homocyclic or heterocyclic radical such as, but not limited to, indolyl, 
dihydroxypyrimidyl or tetra-methylene, 

as well as pharmaceutical^ acceptable addition salts, stereoisomers, mono- 
5 or di-A/-oxides, solvates and/or dihydro- or tetrahydropteridine derivatives 
thereof, are useful for the manufacture of a medicament for the prevention or 
treatment of a TNF-a-related disorder in a mammal, the said disorder being 
selected from the group consisting of: 

- septic or endotoxic shock, 

1 0 - TNF-a- mediated diseases, 

- pathologies and conditions associated with and/or induced by abnormal 
levels of TNF-a occurring in a systemic, localized or particular tissue type 
or location in the body of the mammal, 

- toxic effects of TNF-a and/or anti-cancer chemotherapeutic agents, 

1 5 - injuries after irradiation of a tissue of the mammal by radio-elements, and 

- cachexia. 

The present invention also relates to various processes and methods 
for making the novel ptendine derivatives defined in general formula (I), as 
well as their pharmaceutical^ acceptable salts, N-oxides, solvates, 
20 enantiomers and/or dihydro- and tetrahydroderivatives 

BRIEF DESCRIPTION OF THE DRAWINGS 

Figures 1 and 2 represent alternative schemes for preparing 2,4,6- 
trisubstituted ptendine derivatives used in this invention. 

25 Figure 3 represents a scheme for preparing 2,4,7-trisubstituted 

pteridine derivatives used in this invention. 

Figure 4 represents a scheme for preparing unsymmetrical 2,4,6- 
tnsubstituted pteridines and 2,4,7-trisubstituted pteridines used in this 
invention. 

30 Figure 5 represents a scheme for preparing symmetrical 2,4,6- 

trisubstituted pteridines and 2,4,7-trisubstituted pteridines used in this 
invention. 



12 



DEFINITIONS 

Unless otherwise stated herein, the term " trisubstituted " means that 
three of the carbon atoms being in positions 2, 4 and 6 or, alternatively, in 
positions 2, 4 and 7 of the pteridine ring (according to standard atom 
numbering for the pteridine ring) are substituted with an atom or group other 
than hydrogen. The term " tetrasubstituted " means that all four carbon atoms 
being in positions 2, 4, 6 and 7 of the pteridine ring are substituted with an 
atom or group other than hydrogen. 

As used herein with respect to a substituting radical, and unless 
otherwise stated, the terms " Ci_ 7 alkyl" or " aliphatic saturated hydrocarbon 
radicals with 1 to 7 carbon atoms " means straight and branched chain 
saturated acyclic hydrocarbon monovalent radicals having from 1 to 7 carbon 
atoms such as, for example, methyl, ethyl, propyl, n-butyl, 1-methylethyl 
(isopropyl), 2-methylpropyl (isobutyl), 1,1-dimethylethyl (ter-butyl), 2-methyl- 
butyl, n-pentyl, dimethylpropyl, n-hexyl, 2-methylpentyl, 3-methylpentyl, n- 
heptyl and the like; the term " Cm alkyl " designate the corresponding radicals 
with only 1 to 4 carbon atoms, and so on. 

As used herein with respect to a substituting radical, and unless 
otherwise stated, the term Ci_ 7 alkylene means the divalent hydrocarbon 
radical corresponding to the above defined Ci_ 7 alkyl, such as methylene, 
bis(methylene), tns(methylene), tetramethylene, hexamethylene and the like. 

As used herein with respect to a substituting radical, and unless 
otherwise stated, the terms " Ca-io cydoalkyl" and " cycloaliphatic saturated 
hydrocarbon radical with 3 to 10 carbon atoms " means a monocyclic 
saturated hydrocarbon monovalent radical having from 3 to 10 carbon atoms, 
such as for instance cyclopropyl, cyclobutyl, cyclopentyl, cydohexyl, 
cycloheptyl, cyclooctyl and the like, or a C?-to polycyclic saturated hydrocarbon 
monovalent radical having from 7 to 10 carbon atoms such as, for instance, 
norbomyl, fenchyl, trimethyltricydoheptyl or adamantyl. 

As used herein with respect to a substituting radical, and unless 
otherwise stated, the term " C3.10 cydoalkylene" means the divalent 
hydrocarbon radical corresponding to the above defined C3-10 cycloalkyl. 



13 



As used herein with respect to a substituting radical, and unless 
otherwise stated, the terms " aryl " and "aromatic substituent " are 
interchangeable and designate any mono- or polyaromatic monovalent 
hydrocarbon radical having from 6 up to 30 carbon atoms such as but not 
5 limited to phenyl, naphthyl, anthracenyl, adamantyl, phenantracyl, 
fluoranthenyl, chrysenyl, pyrenyl, biphenylyl, terphenyl, picenyl and the like, 
including spiro hydrocarbon radicals and fused benzo - C M cycloalkyl radicals 
(the latter being as defined above) such as, for instance, indanyl, 1,2,3,4- 
tetrahydronaphtalenyl, fluorenyl and the like. 
10 As used herein with respect to a substituting radical such as the 

combination of R3 and R4, and unless otherwise stated, the term " homo- 
cyclic" means a mono- or polycyclic, saturated or mono-unsaturated or 
polyunsaturated hydrocarbon radical having from 4 up to 15 carbon atoms but 
including no heteroatom in the said ring. 
15 As used herein with respect to a substituting radical, and unless 

otherwise stated, the term " heterocyclic " means a mono- or polycyclic, 
saturated or mono-unsaturated or polyunsaturated monovalent hydrocarbon 
radical having from 2 up to 15 carbon atoms and including one or more 
heteroatoms in a 3 to 10 membered ring (and optionally one or more 
20 heteroatoms attached to one or more carbon atoms of said ring, for instance 
in the form of a carbonyl or thiocarbonyl group) and/or to one or more 
heteroatoms of said ring, for instance in the form of a sulfone, sulfoxide, N- 
oxide, phosphate, phosphonate or selenium oxide, each said heteroatom 
being independently selected from the group consisting of nitrogen, oxygen, 
25 sulfur, selenium and phosphorus, including benzo-fused heterocyclic radicals, 
such as but not limited to dibenzothiophenvl, dibenzofuranyl, oxazolinyl, 
oxazolonyl, azaindolyl, azolonyl, thiazolinyl, thiazolonyl, thiazolidinyl, thiazanyl, 
pyrimidonyl, thiopyrimidonyl, thiamorpholinyl, azlactonyl, naphtindazolyl, 
naphtindolyl, naphtothiazolyl, naphtothbxolyl, naphtoxindolyl. naphtotriazolyl, 
30 naphtopyranyl, oxabicycloheptyl, azabenzimidazolyl, azacydoheptyl, aza- 
cyclooctyl, azacyclononyl, azabicyclononyl, tetrahydroforyl, tetrahydropyranyl, 
tetrahydropyronyl, tetrahydroquinoleinyl, tetrahydrothienyl and dioxide thereof, 
dihydrothienyl dioxide, dioxindolyl, dioxinyl, dioxenyl, dioxazinyl, thioxanyl, 



14 



thioxolyl, thiourazolyl, thiotnazolyl, thiopyranyl, thiopyranyl, coumarinyl, 
quinoleinyl, oxyquinoleinyl, quinuclidinyl, xanthinyl, dihydro-pyranyl, 
benzodihydrofuryl, benzothiopyronyl, benzothiopyranyl, benzoxazinyl, 
benzoxazolyl, benzodioxolyl, benzodioxanyl, benzothiadiazolyl, benzotriazinyl, 
benzothiazolyl, benzoxazolyl, phenothioxinyl, phenothiazolyl, phenothienyl 
(benzothiofuranyl), phenopyronyl, phenoxazolyl, pyridinyl, dihydropyridinyl, 
tetrahydropyridinyl, piperidinyl, morpholinyl, thiomorpholinyl, pyrazinyl, 
pyrimidinyl, pyridazinyl, triazinyl, tetrazinyl, triazolyl, benzotriazolyl, tetrazolyl, 
imidazolyl, pyrazolyl, thiazolyl, thiadiazolyl, isothiazolyl, oxazolyl, oxadiazolyl, 
pyrrolyl, furyl, dihydrofuryl, furoyl, hydantoinyl, dioxolanyl, dioxolyl, dithianyl, 
dithienyl, dithifnyl, thienyl, indolyl, indazolyl, benzofuryl, quinolyl, quinazolinyl, 
quinoxalinyl, carbazolyl, phenoxazinyl, phenothiazinyl, xanthenyl, purinyl, 
benzothienyl, naphtothienyl, thianthrenyl, pyranyl, pyronyl, benzopyronyl, 
isobenzofuranyl, chromenyl, phenoxathiinyl, indolizinyl, quinolizinyl, 
isoqumolyl, phthalazinyl, naphthiridinyl, cinnolinyl, pteridinyl, carbolinyl, 
acridinyl, perimidinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, imidazolinyl, 
imidazolidinyl, benzimidazolyl, pyrazolinyl, pyrazolidinyl, pyrrolinyl, pyrrolidinyl, 
piperazinyl, uridinyl, thymidinyl, cytidmyl, azirinyl, azindinyl, diazirinyl, 
diaziridinyl, oxiranyl, oxaziridinyl, dioxiranyl, thiiranyl, azetyl, dihydroazetyl, 
azetidinyl, oxetyl, oxetanyl, thietyl, thietanyl, diazabicyclooctyl, diazetyl, 
diaziridinonyl, diaziridinethionyl, chromanyl, chromanonyl, thiochromanyl, 
thiochromanonyl, thiochromenyl, benzofuranyl, benzisothiazolyl, benzocarb- 
azolyl, benzochromonyl, benzisoalloxazinyl, benzocoumarinyl, thiocoumarinyl, 
phenometoxazinyl, phenoparoxazinyl, phentriazinyl, thiodiazinyt, thiodiazolyl, 
indoxyl, thio-indoxyl, benzodiazinyl (e.g. phtalazinyl), phtalidyl, phtalimidinyl, 
phtalazonyl, alloxazinyl, dibenzopyronyl (i.e. xanthonyl), xanthionyl, isatyl, 
isopyrazolyl, isopyrazolonyl, urazolyl, urazinyl, uretinyl, uretidinyl, succinyl, 
succimmido, benzylsultimyl, benzylsultamyl and the like, including all possible 
isomeric fonms thereof, wherein each carbon atom of the said ring may be 
substituted with a substituent selected from the group consisting of halogen, 
nitro, C1-7 alkyl (optionally containing one or more functions or radicals 
selected from the group consisting of carbonyl (oxo), alcohol (hydroxyl), ether 
(alkoxy), acetal, amino, imino, oximino, alkyloximino, amino-atid, cyano, 



15 

carboxylic acid ester or amide, nitro, thio C1.7 alkyl, thio C3-10 cycloalkyl. C1-7 
alkylamino, cydoalkylamino, alkenylamino, cycloalkenylamino, alkynylamino, 
arylamino, arylalkylamino, hydroxyalkylamino, mercaptoalkylamino, 
heterocyclic amino, hydrazino, alkylhydrazino, phenylhydrazino, sulfonyl, 
5 sulfonamide and halogen), C3.7 alkenyl, C2-7 alkynyl, halo C1.7 alkyl, C3.10 
cycloalkyl. aryl, arylalkyl. alkylaryl, alkylacyl. arylacyl, hydroxyl, amino, C1-7 
alkylamino, cydoalkylamino, alkenylamino, cydo-alkenylamino. alkynylamino, 
arylamino, arylalkylamino, hydroxyalkylamino, mercaptoalkylamino, hetero- 
cyclic amino, hydrazino, alkylhydrazino, phenylhydrazino, sulfhydryl, C1-7 

10 alkoxy, C3.10 cycloalkoxy, aryloxy, arylalkyloxy. oxyheterocydic, heterocydic- 
substituted alkyloxy, thio C1-7 alkyl, thio C3-10 cydoalkyl, thioaryl, 
thioheterocydic, arylalkylthio, heterocydic-substituted alkylthio, formyl, 
hydroxylamino, cyano, carboxylic add or esters or thioesters or amides 
thereof, thiocarboxylic add or esters or thioesters or amides thereof; 

15 depending upon the number of unsaturations in the 3 to 10 membered ring, 
heterocydic radicals may be sub-divided into heteroaromatic (or " heteroaryl") 
radicals and non-aromatic heterocydic radicals, when a heteroatom of the 
said non-aromatic heterocydic radical is nitrogen, the latter may be 
substituted with a substituent seleded from the group consisting of C1.7 alkyl, 

20 Ca.10 cydoalkyl, aryl, arylalkyl and alkylaryl. 

As used herein with resped to a substituting radical, and unless 
otherwise stated, the terms " C1-7 alkoxy ", "Ca-io cydoalkoxy ", " aryloxy", 
"arylalkyloxy ", " oxyheterocydic ", "thio C1.7 alkyl", " thio C3.10 cydoalkyl ", 
"arylthio ", " arylalkylthio " and " thioheterocydic" refer to substituents 

25 wherein a C1-7 alkyl radical, respectively a Cwo cydoalkyl, aryl, arylalkyl or 
heterocydic radical (each of them such as defined herein), are attached to an 
oxygen atom or a sulfur atom through a single bond, such as but not limited to 
methoxy, ethoxy, propoxy. butoxy, thioethyl, thiomethyl, phenyloxy, benzyloxy, 
mercaptobenzyl, cresoxy and the like. 

30 As used herein with resped to a substituting atom, and unless 

otherwise stated, the term halogen means any atom selected from the group 
consisting of fluorine, chlorine, bromine and iodine. 



16 



As used herein with respect to a substituting radical, and unless 
otherwise stated, the term " halo C1.7 alkyl " means a C1.7 alkyl radical (such 
as above defined) in which one or more hydrogen atoms are independently 
replaced by one or more halogens (preferably fluorine, chlorine or bromine), 
5 such as but not limited to difluoromethyl, trifluoromethyl, trifluoroethyl, 
octafluoropentyl, dodecafluoroheptyl, dichloromethyl and the like; the term 
"halo C1.4 alkyl " designate the corresponding radical with only 1 to 4 carbon 
atoms, and so on. 

As used herein with respect to a substituting radical, and unless 
10 otherwise stated, the terms " C2-7 alkenyl " and " aliphatic unsaturated 
hydrocarbon radical with 2 to 7 carbon atoms " are interchangeable and 
designate a straight and branched acyclic hydrocarbon monovalent radical 
having one or more ethylenical unsaturations and having from 2 to 7 carbon 
atoms such as, for example, vinyl, 2-propenyl. 3-butenyl, 2-butenyl, 2- 
15 pentenyl, 3-pentenyl, 3-methyl-2-butenyl, 3-hexenyl, 2-hexenyl, 2-heptenyl, 
butadienyl, pentadienyl, hexadienyl, heptadienyl, heptatrienyl and the like, 
including all possible isomers thereof; the term " C3-7 alkenyl " designate the 
corresponding radical with only 3 to 7 carbon atoms, and so on. 

As used herein with respect to a substituting radical, and unless 

20 otherwise stated, the terms " C3-10 cycloalkenyl" and " cycloaliphatic 
unsaturated hydrocarbon radical with 3 to 10 carbon atoms " are 
interchangeable and mean a monocyclic mono- or polyunsaturated 
hydrocarbon monovalent radical having from 3 to 8 carbon atoms, such as for 
instance cyclopropenyl, cyclobutenyl, cyclopentenyl, cydopentadienyl, 

25 cyclohexenyl, cyclohexadienyl, cycloheptenyl, cycloheptadienyl, cydohepta- 
trienyl, cyclooctenyl, cyclooctadienyl and the like, or a C7-10 polycydic mono- 
or polyunsaturated hydrocarbon mono-valent radical having from 7 to 10 
carbon atoms such as dicydopentadienyl, fenchenyl (induding all isomers 
thereof, such as a-pinolenyl), bicydo[2.2 1]hept-2-enyl, bicyclo[2.2.1]hepta- 

30 2,5-dienyl, cydo-fenchenyl and the like. 

As used herein with resped to a substituting radical, and unless 
otherwise stated, the term " C2-7 alkynyl " defines straight and branched chain 
hydrocarbon radicals containing one or more triple bonds and having from 2 to 



17 



20 carbon atoms such as, for example, acetylenyl, 2-propynyl, 3-butynyl, 2- 
butynyl, 2-pentynyl, 3-pentynyl, 3-methyl-2-butynyl, 3-hexynyl, 2-hexynyl and 
the like and all possible isomers thereof. 

As used herein with respect to a substituting radical, and unless 
5 otherwise stated, the terms " arylalkyl " and "heterocyclic-substituted alkyl" 
refer to an aliphatic saturated hydrocarbon monovalent radical, preferably a 
C1-7 alkyl or a C3.10 cycloalkyl such as defined above, onto which an aryl 
radical or respectively a heterocyclic radical (such as defined above) is 
already bonded, such as but not limited to benzyl, pyridylmethyl, pyridylethyl, 
10 2-(2-pyridyl)isopropyl, oxazolylbutyl, 2-thienylmethyl and 2-furylmethyl. 

As used herein with respect to a substituting radical, and unless 
otherwise stated, the term " alkylaryl " and "alkyl-substituted heterocyclic" 
refer to an aryl radical or respectively a heterocyclic radical (such as defined 
above) onto which is (are) already bonded one or more aliphatic saturated 
15 hydrocarbon monovalent radicals, preferably C1.7 alkyl radicals or C3.10 
cycloalkyl radicals as defined above such as, but not limited to, o-toluyl, m- 
toluyl, p-toluyl, mesityl and 2,4,6-trimethylphenyl. 

As used herein with respect to a substituting radical, and unless 
otherwise stated, the terms " alkylamino ", "cycloalkylamino ", "alkenyl- 
20 . amino", " cycloalkenylamino " , " arylamino ", "arylalkylamino", "heterocyclic 
amino " , " hydroxyalkylamiho ", "mercaptoalkylamino " and " alkynylamino" 
mean that respectively one (thus monosubstituted amino) or even two (thus 
disubstituted amino) Cur alkyl, C^o cycloalkyl, C2-7 alkenyl, C3-10 cycloalkenyl, 
aryl, arylalkyl, heterocyclic, mono- or polyhydroxy C1-7 alkyl, mono- or 
25 polymercapto C1.7 alkyl or C2-7 alkynyl radical(s) (each of them as defined 
herein, respectively) is/are attached to a nitrogen atom through a single bond 
or, in the case of heterocyclic, include a nitrogen atom, such as but not limited 
to, anilino, benzylamino, methylamino, dimethylamino, ethylamino, 
diethylamino, isopropylamino, propenylamino, n-butylamino, ter-butylamino, 
30 dibutylamino, morpholino-alkylamino, morpholinyi, piperidinyl, piperazinyl, 
hydroxymethylamino, p-hydroxyethylamino and ethynylamino; this definition 
also includes mixed disubstituted amino radicals wherein the nitrogen atom is 
attached to two such radicals belonging to two different sub-set of radicals, 



18 



e.g an alkyl radical and an alkenyl radical, or to two different radicals within 
the same sub-set of radicals, e.g. methylethylammo; the term " C3.7 alkyl- 
amino" designates the corresponding radical with only 3 to 7 carbon atoms in 
the alkyl group(s) attached to nitrogen, for instance di-isopropylamino, and so 
5 on; among disubstituted amino radicals, symetrically substituted are usually 
preferred and more easily accessible. 

As used herein with respect to a substituting radical, and unless 
otherwise stated, the terms "(thio)carboxylic add ester " , " (thio)carboxylic 
acid thioester " and " (thio)carboxylic add amide" refer to radicals wherein the 
10 carboxyl or thiocarboxyl group is directly attached to the pteridine ring (e.g. in 
the 6- and/or 7-position) and wherein said carboxyl or thiocarboxyl group is 
bonded to the hydrocarbonyl residue of an alcohol, a thiol, a polyol, a phenol, 
a thiophenol, a primary or secondary amine, a polyamine, an amino-alcohol or 
ammonia, the said hydrocarbonyl residue being selected from the group 

15 consisting of alkyl, alkenyl, alkynyl, cydoalkyl, cydoalkenyl, aryl, arylalkyl, 
alkylaryl, alkylamino, cydoalkylamino, alkenylamino, cydoalkenylamino, 
arylamino, arylalkylamino, heterocydic amino, hydroxyalkylamino, mercapto- 
alkylamino or alkynylamino (such as above defined, respectively). 

As used herein with respect to a substituting radical, and unless 

20 otherwise stated, the term " amino-add " refers to a radical derived from a 
molecule having the chemical formula H 2 N-CHR--COOH, wherein R is the 
side group of atoms characterizing the amino-add type; said molecule may be 
one of the 20 naturally-occurring amino-acids or any similar non naturally- 
occurring amino-add. 

25 As used herein and unless otherwise stated, the term " stereoisomer " 
refers to all possible different isomeric as well as conformational forms which 
the pteridine derivatives having the general formula (I) may possess, in 
particular all possible stereochemical^ and conformationally isomeric forms, 
all diastereomers, enantiomers and/or contemners of the basic molecular 

30 structure. Some compounds of the present invention may exist in different 
tautomeric forms, all of the latter being induded within the scope of the 
present invention. 



19 

As used herein and unless otherwise stated, the term " enantiomer " 
means each individual optically active form of a compound of the invention, 
having an optical purity or enantiomeric excess (as determined by methods 
standard in the art) of at least 80% (i.e at least 90% of one enantiomer and at 
most 10% of the other enantiomer), preferably at least 90% and more 
preferably at least 98%. 

As used herein and unless otherwise stated, the term " solvate " 
includes any combination which may be formed by a pteridine derivative of 
this invention with a suitable inorganic solvent (e.g. hydrates) or organic 
solvent, such as but not limited to alcohols, ketones, esters and the like. 

As used herein and unless otherwise stated, the terms " dihydro- 
pteridine derivative " and " tetrahydropteridine derivative " refer to the 
hydrogenation products of the pteridine denvatives having the general formula 
(I), i.e. derivatives wherein two hydrogen atoms are present in positions 5 and 
6, or 7 and 8, of the ptendine nng, or respectively wherein four hydrogen 
atoms are present in positions 5, 6, 7 and 8 of the said ring; such 
hydrogenated derivatives are easily accessible from the pteridine derivatives 
of formula (I) by using hydrogenation methods well known in the art. 

DETAILED DESCRIPTION OF THE INVENTION 

A main object of the invention is to provide a treatment for a class of TNF-a- 
related disorders in a mammal, the said disorders being selected from the group 
consisting of: 

- septic or endotoxic shock, 

- TNF-<j-mediated diseases, 

- pathologies and conditions associated with and/or induced by abnormal 
levels of TNF-a occurring in a systemic, localized or particular tissue type 
or location in the body of the mammal, 

- toxic effects of TNF-a and/or other anti-cancer chemotherapeutic agents, 

- injunes after irradiation of a tissue of the mammal by radio-elements, and 

- cachexia. 



20 



This is achieved by manufacturing a medicament, or a pharmaceutical 
composition, including a pteridine derivative having the above mentioned 
general formula (I) as a biologically active ingredient 

According to the invention, the active pteridine derivatives are as 
defined in the general formula (I), wherein each of the substituents X, Z, R 1t 
R2. R3 and R4 may correspond to any of the definitions given above (and, 
when X includes sulfur, wherein m may be 0, 1 or 2), in particular with any of 
the individual meanings (such as illustrated above) of generic terms such as, 
but not limited to, "C.7 alkyl ", " C 27 alkenyl ", - C 2 - 7 alkynyl ", " aryl ", 
"alkylaryl ", "arylalkyl ", "alkylamino", " cycloalkylamino ", "alkenylamino 
"alkynylamino", "arylammo", " arylalkylamino ", " C1.7 alkoxy", " C3.10 
cycloalkoxy ", "thio C1.7 alkyl ", " thio C3.10 cycloalkyl ", - halo C1-7 alkyl ", 
"amino-add " and the like In particular, the pteridine derivatives wherein R2 is 
C3.7 alkylamino and/or wherein R4 is a heterocyclic radical other than 
piperidinyl, morpholinyl or pyrrolidinyl (i.e. R4 is for instance selected from the 
group consisting of dibenzothiophenyl, dibenzofuranyl, oxazolinyl, oxazolonyl, 
azaindolyl, azolonyl, thiazolinyl, thiazolonyl, thiazolidinyl, thiazanyl, 
pyrimidonyl, thiopyrimidonyl, azlactonyl, naphtindazolyl, naphtindolyl, 
naphtothiazolyl, naphtothioxolyl, naphtoxindolyl, naphtotriazolyl, naphto- 
pyranyl, oxabicydoheptyl, azabenzimidazolyl, azacycloheptyl, azacydooctyl 
dihydrothienyl dioxide, dioxindolyl, dioxinyl, dioxenyl, dioxazinyl, thioxanyl, 
thioxolyl, thiourazolyl, thiotriazolyl, thiopyranyl, thiopyronyl, coumarinyl, quino- 
leinyl, oxyquinoleinyl, quinudidinyl, xanthinyl, dihydropyranyl, benzodihydro- 
furyl, benzothiopyronyl, benzothiopyranyl, benzoxazinyl, benzoxazolyl, 
benzodioxolyl, benzodioxanyl, benzothiadiazolyl, benzotriazinyl, 
benzothiazolyl, benzoxazolyl, phenothioxinyl, phenothiazolyl, phenothienyl, 
phenopyronyl, phenoxazolyl, pyridinyl, dihydropyridinyl, tetrahydropyridinyl, 
thiomorpholinyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, tetrazinyl, triazolyl, 
benzotriazolyl, tetrazolyl, imidazolyl, pyrazolyl, thiazolyl, thiadiazolyl, 
isothiazolyl, oxazolyl, oxadiazolyl, pyrrolyl, furyl, dihydrofuryl, furoyl, 
hydantoinyl, dioxolanyl, dioxolyl, dithianyl, dithienyl, dithiinyl, thienyl, indolyl, 
indazolyl, benzofuryl, quinolyl, quinazolinyl, quinoxalinyl, azacydononyl, 
azabicydononyl, tetrahydrofuryl, tetrahydropyranyl, tetrahydropyronyl, 



21 



tetrahydroquinoleinyl, tetrahydrothienyl and dioxide thereof, carbazolyl, 
phenoxazinyl, phenothiazinyl, xanthenyl, purinyl, benzothienyl, naphtothienyl, 
thianthrenyl, pyranyl, pyronyl, benzopyronyl, isobenzofuranyl, chromenyl, 
phenoxathiinyl, indolizinyl, quinolizinyl, isoquinolyl, phthalazinyl, naphthiridinyl, 
cinnolinyl, pteridinyl, carbolinyl, acridinyl, perimidinyl, phenarrthrolinyl, 
phenazinyl, phenothiazinyl, imidazolinyl, imidazolidinyl, benzimidazolyl, 
pyrazolinyl, pyrazolidinyl, pyrrolinyl, piperazinyl, uridinyl, thymidinyl, cytidinyl. 
azirinyl, aziridinyl, diazirinyl, diaziridinyl, oxiranyl, oxaziridinyl, dioxiranyl, 
thiiranyl, azetyl, dihydroazetyl, azetidinyl, oxetyl, oxetanyl, thietyl, thietanyl, 
diazabicyclooctyl, diazetyl, diaziridinonyl, diaziridinethionyl, chromanyl, 
chromanonyl, thiochromanyl, thiochromanonyl, thiochromenyl, benzofuranyl, 
benzisothiazolyl, benzocarbazolyl, benzochromonyl, benzisoalloxazinyl, 
benzocoumarinyl, thiocoumarinyl, phenometoxazinyl, phenoparoxazinyl, 
phentriazinyl, thiodiazinyl, thiodiazolyl, indoxyl, thioindoxyl, benzodiazinyl, 
phtalidyl, phtalimidinyl, phtalazonyl, alloxazinyl, dibenzopyronyl, xanthionyl, 
isatyl, isopyrazolyl, isopyrazolonyl, urazolyl, urazinyl, uretinyl, uretidinyl, 
succinyl, succinimido, benzylsultimyl and benzylsultamyl) are novel 
compounds which were never suggested for use in medicine. 

When a mixture of enantiomers of a pteridine derivative having the 
general formula (I) according to the invention is obtained during synthesis, the 
said mixture may be separated by means and methods standard in the art, 
e.g. liquid chromatography using one or more suitable chiral stationary 
phases. The latter include, for example, polysaccharides, in particular 
cellulose or amylose derivatives. Commercially available polysaccharide- 
based chiral stationary phases suitable for this purpose are ChiralCel™ CA, 
OA, OB, OC, OD, OF, OG, OJ and OK, and Chiralpak™ AD, AS, OP(+) and 
OT(+). Appropriate eluents or mobile phases for use in combination with said 
polysaccharide-based chiral stationary phases are hydrocarbons such as 
hexane and the like, optionally admixed with an alcohol such as ethanol, 
isopropanol and the like. The above mixture of enantiomers may alternatively 
be separated by forming diastereoisomers, followed by separation of the 
diastereoisomers, e.g. by differential crystallization or chromatography. The 



22 



resolving agent may be cleaved from the separated diastereoisomers, e.g. by 
treatment with acids or bases, in order to generate the pure enantiomers of 
the compounds of the invention. 

Some preferred pteridine derivatives having the general formula (I) 
5 according to the invention are more specifically illustrated in the following 
examples For instance, useful pteridine species disclosed below include 
these wherein: 

- Ri is selected from the group consisting of methyl, ethyl, isopropyl, pentyl 
and benzyl, and/or 

10 - • R 2 is amino, and/or 

- R4 is hydrogen or methoxy, and/or 

- R3 is 3-thienyl or 2-thienyl or a phenyl group with one or more substituents 
(in the latter case, such substituents are preferably each independently 
selected from the group consisting of fluoro, methoxy, ethoxy, 

15 trifluoromethyl, dimethyl-amino, chloro. cyano, methyl, ethyl, 
carboxymethyl, methylthio, dimethylcarboxamido, diethylcarboxamido and 
methylcarboxylate, and/or 

- X is a sulfur atom (i.e. m is 0) or an oxygen atom, or 

- X is NZ, wherein Z is selected from the group consisting of hydrogen, 
20 methyl, ethyl, isopropyl and benzyl, or NZ together with R, forms a radical 

selected from the group consisting of hydroxylamino, tetrahydropyndinyl, 
morpholinyl, piperidinyl, piperazinyl, N-methylpiperazinyl. 1,2,4-triazolyl 
and pyrrolidinyl 

The present invention further provides processes and methods for making 
25 the pteridine derivatives having the general formula (I). As a general rule, the 
preparation of these compounds is based on the principle that, starting from a 
suitable pteridine precursor, each of the substituents XRi , R 2 , R3 and R4may 
be introduced separately (except, of course, when R3 together with R4 forms a 
homocyclic or heterocyclic radical) without adversely influencing the presence 
30 of one or more substituents already introduced at other positions on the 
pteridine ring or the capacity to introduce further substituents later on. 

Methods of manufacture have been developed by the present inventors 
which may be used alternatively to, or may be combined with, the methods of 



23 



synthesis already known in the art of pteridine derivatives (depending upon 
the targeted final compound). For instance, methods for simultaneously 
introducing R3 and R4 in the form of a homocyclic or heterocyclic radical at 
positions 6 and 7 of the pteridine ring are already known from U.S. Patent No. 
5 2,581 ,889. The synthesis of mono- and di-/V-oxides of the pteridine derivatives 
of this invention can easily be achieved by treating the said derivatives with an 
oxidizing agent such as, but not limited to, hydrogen peroxide (e.g. in the 
presence of acetic acid) or a peracid such as chloroperbenzoic acid. Dihydro- 
and tetrahydropteridine derivatives of this invention can easily be obtained by 

10 catalytic hydrogenation of the corresponding pteridine derivatives, e.g. by 
placing the latter in a hydrogen atmosphere in the presence of platinum oxide 
or platinum. The methods for making the pteridine derivatives of the present 
invention will now be explained in more details by reference to the appended 
figures 1 to 5 wherein, uniess otherwise stated hereinafter, each of the 

1 5 substituting groups or atoms X, Z, R 1( R 2l R 3 and R4 is as defined in formula (I) 
of the summary of the invention and, more specifically, may correspond to any 
of the individual meanings disclosed above. The same manufacturing 
methods may also be applied, if need be, while starting from pteridine 
derivatives which are already known in the art. In the description of the 

20 reaction steps involved in each figure, reference is made to the use of certain 
catalysts and/or certain types of solvents. It should be understood that each 
catalyst mentioned should be used in a catalytic amount well known to the 
skilled person with respect to the type of reaction involved. Solvents that may 
be used in the following reaction steps include various kinds of organic 

25 solvents such as protic solvents, polar ^p^otic solvents and non-polar solvents 
as well as aqueous solvents which are inert under the relevant reaction 
conditions. More specific examples include aromatic hydrocarbons, 
chlorinated hydrocarbons, ethers, aliphatic hydrocarbons, alcohols, esters, 
ketones, amides, water or mixtures thereof, as well as supercritical solvents 

30 such as carbon dioxide (while performing the reaction under supercritical 
conditions). The suitable reaction temperature and pressure conditions 
applicable to each kind of reaction step will not be detailed herein but do not 
depart from the relevant conditions already known to the skilled person with 



24 



respect to the type of reaction involved and the type of solvent used (in 
particular its boiling point). 

Figure 1 represents a scheme for the preparation of 2,4,6-trisubstituted 
pteridines with various R 2 and R3 substituents in the 2- and 6-positions of the 
pteridine ring. In the first step (a), a chloropyrimidine 1, wherein R 2 may be 
inter alia amino, alkylamino, arylamino, alkoxy, aryloxy, mercaptoalkyl, or 
mercaptoaryl, is reacted with an appropriate nucleophile R^H, the said 
nucleophile being selected from the group consisting of alcohols (e.g. 
methanol, ethanol, isopropanol or benzylalcohol), thiols, primary amines and 
secondary amines wherein R t may be inter alia alkyl, cycloalkyl, aryl, alkylaryl, 
heteroaryl or alkylheteroaryl. Introduction of a nitroso group into the pyrimidine 
intermediate 2 occurs in step (b) under acidic aqueous conditions in the 
presence of sodium nitrite NaN0 2 . Reduction of the nitroso functionality of the 
pynmidine intermediate 3 into a free amino group in intermediate 4 is then 
effected in step (c) by means of reducing agents (such as Na2S 2 0 4 or 
(NH 4 ) 2 S) in water, or catalytically (Pt/H 2 ) in the presence of a protic solvent In 
step (d), ring closure is performed by treating the diaminopyrimidine 4 with 
glyoxal in order to form a pteridine ring. In step (e), the nitrogen atom at 
position 8 of the pteridine ring of compound 5 is oxidized, e.g. using Kb0 2 
under acidic conditions. In step (f), a chlorine atom is regioselectively 
introduced on the 6 position of the pteridine ring of compound 6 by treatment 
with a carboxylic acid choride such as acetyl chloride under acidic conditions. 
Then in step (g) the 6-chlorosubstituted pteridine 7 is reacted with a boronic 
acid having the general formula R3B(OH) 2 , wherein R3 may be alkyl, 
cycloalkyl, aryl or heteroaryl, under basic conditions (such as in the presence 
of an aqueous alcaline solution) and a palladium based catalyst, thus yielding 
the desired derivative 8 of the present invention. 

Figure 2 represents a scheme for the preparation of 2,4,6-trisubstituted 
pteridines with various R 2 and R3 substituents in the 2- and 6-positions of the 
ptendine ring. In step (a), a nitroso group is introduced in position 5 of the 
pyrimidine ring of a compound 9, wherein R 2 may be inter alia amino, 
alkylamino or arylamino, using sodium nitrite under aqueous acidic conditions. 
Reduction of the nitroso group of compound 10 in step (b) is achieved either 



25 



catalytically (Pt/H 2 ) in the presence of a protic solvent, or chemically using 
sodium dithionite or ammonium sulfide in the presence of water. Then in the 
next step (c), the condensation of the diaminopynmidine 11 with an a- 
ketoaldoxime bearing the group R3, wherein R3 may be alkyl, cycloalkyl, aryl 
5 or heteroaryl, under acidic conditions in the presence of a protic solvent such 
as methanol, regioselectively yields the 6-substituted pteridine derivative 12. 
Activation of the hydroxyl group of the tautomeric form of 12 by a nucleophilic 
displacement reaction occurs by preparing the 4-R1 ,2,4)-triazolyl] pteridine 
derivative 13, using POCI 3 or 4-chlorophenyl phosphorodichloridate, and 

10 1,2,4-triazole in the presence of e.g. pyridine as solvent. If R 2 is a free amino 
group, protection of R 2 e.g. by means of an acetyl group may be necessary 
before carrying out the reaction of step (d), followed by a deprotection of the 
acetyl group during the nucleophilic displacement reaction. The nucleophilic 
substitution in step (e) may be performed, e.g. in the presence of 1,4-dioxane 

15 as a solvent, by mixing the pteridine derivative 13 at room temperature with an 
appropriate nucleophile RiXH, the said nudeophile being selected from the 
group consisting of alcohols (e.g. methanol, ethanol, isopropanol or 
benzylalcohol), thiols, primary amines and secondary amines wherein R t may 
be inter alia alkyl, cycloalkyl, aryl, alkylaryl, heteroaryl or alkylheteroaryl, thus 

20 yielding the desired derivative 14 of the present invention. 

Figure 3 represents a scheme for the preparation of 2,4,7-trisubstrtuted 
pteridines with various R 2 and R« substituents in the 2- and 7-positions of the 
pteridine ring. In reaction step (a), a tetraaminopyrimidine 15, wherein R 2 may 
be inter alia amino, alkylamino or arylamino, is reacted with inter alia an 

25 alkylglyoxal, arylglyoxal, alkylarytglyoxal, heteroarylglyoxal or alkyl 
heteroarylglyoxat in water under basic conditions, yielding the 7-substituted 
pteridine derivative 16 wherein R4 may be inter alia alkyl, cycloalkyl, aryl, 
heteroaryl, alkylaryl or alkylheteroaryl. In step (b), hydrolysis under reflux in 
the presence of sodium hydroxide 1N yielded the 4-hydroxy tautomer of 

30 pteridine derivative 17. In step (c), a 1 ,2,4-triazolyl group is introduced in 
position 4 by reacting 17 with triazole in the presence of 4-chlorophenyl 
phosphorodichloridate and dry pyridine. Finally in reaction step (d). the 1,2,4- 
triazolyl group at position 4 of the pteridine derivative 18 is displaced by an 



26 



appropriate nudeophie RiXH, the said nucleophile being selected from the 
group consisting of alcohols (e.g. methanol, ethanol, isopropanol or 
benzylalcohol), thiols, priman/ amines and secondary amines wherein Ri may 
be inter alia alkyl, cycloalkyl, aryl, alkylaryl, heteroaryl or alkylheteroaryl, in the 
5 presence of a polar aprotc or protic solvent, thus yielding the desired 
derivative 19 of the present invention. 

Figure 4 represents a scheme for the synthesis of unsymmetrical 2,4,6- 
trisubstituted and 2,4,7-tnsubstituted pteridine derivatives with various R 2 , R 3 
and R4 substituents in the 2-, 6- and 7-positions of the pteridine ring. In step 
10 (a), the thiol function on 2-mercapto-4,6-diaminopyrimidine is methylated, e.g. 
by reaction with methyl iodide in the presence of a solvent such as ethanol, in 
order to yield 2-thiomethyl-4,6-diaminopyrimidine. Introduction of a nitroso 
group in the 5-position of the pyrimidine ring is then achieved in step (b) by 
using sodium nitrite under aqueous acidic conditions. In the next step (c), the 
15 methylthio group in the 2-position is exchanged for a group R 2 , wherein R 2 
may be inter alia alkoxy, aryloxy, cycloalkyloxy, heteroaryloxy, mercaptoalkyl, 
mercaptoaryl, mercaptocyclo-alkyl or mercaptoheteroaryl by reaction with an 
appropriate nucleophile. Reduction of the nitroso functionality is then achieved 
in step (d) either catalytically {PVH2) in the presence of a protic solvent or 
20 chemically using sodium dithionite or ammonium sulfide in the presence of 
water. Then in the next step (e), the condensation of the 2-substituted-4,5,6- 
triaminopyrimidine with an a-ketoaldoxime bearing the group R3, wherein R3 
may be inter alia alkyl, cycloalkyl, aryl or heteroaryl, under acidic conditions in 
the presence of a solvent such as methanol, regioselectively yields a 2,6- 
25 substttuted-4-aminoptendine derivative. The corresponding 2,7-substituted-4- 
ammopteridine can be obtained, according to step (i), by reaction of the 2- 
substituted-4,5,6-triaminopyrimidine with a glyoxal bearing the group R4, 
wherein R4 may be inter alia alkyl, cycloalkyl, aryl or heteroaryl. According to 
step (f), acidic or basic hydrolysis of the amino group at position 4 of the 
30 pteridine ring, performed on the derivative from step (e) or (i), yields the 
corresponding 4-oxopteridine derivative. In step (g), the hydroxyl group of the 
tautomeric form of the latter is activated by nucleophilic displacement, e.g. by 
preparing the 4-[(1,2,4)-triazolyl] pteridine derivative Finally, the nucleophilic 



27 



displacement in step (h) is performed by mixing the said 4-triazolyl pteridine 
derivative with the appropriate nucleophile RiXH, wherein Ri may be inter alia 
alkyl, cydoalkyl, aryl,' heteroaryl, alkylaryl, alkylheteroaryl. 

Figure 5 represents a scheme for the synthesis of symmetrical 2,4,6- 
5 ^'substituted and 2,4,7-trisubstituted pteridine derivatives with various R 2 , R3 
and R4 substituents in the 2- 6- and 7-positions of the pteridine ring. In step 
(a), the pyrimidine ring is nitrated in position 5 under strongly acidic conditions 
(HNO3, H2SO4). Then in step (b) both hydroxyl groups (from the tautomeric 
form) are converted to chloro groups by treatment with a chlorinating agent 
10 such as POCI3 or SOCI 2 . Both chloro substituents are then displaced in step 
(c) with an appropriate nucleophile RiXH, wherein R f may be inter alia alkyl, 
cydoalkyl, aryl, heteroaryl, alkylaryl, alkylheteroaryl. The nitro group is 
reduced in step (d) to an amino group by treatment with a redudng agent (e.g. 
Pt/H 2 ). Finally, reaction of the 2,4-substituted-5,6-diaminopyrimidine from step 

15 (d) with an a-ketoaldoxime bearing the group R3, wherein R3 may be inter alia 
alkyl, cydoalkyl, aryl or heteroaryl, regioselectively yields the desired 2,4,6- 
trisubstituted pteridine derivative in step (e). Alternatively, readion of the 2,4- 
substituted-5,6-diaminopyrimidine from step (d) with a glyoxal bearing the 
group R4, wherein R4 may be inter alia alkyl, cydoalkyl, aryl or heteroaryl, 

20 yields the desired 2,4,7-trisubstituted pteridine derivative in step (f). 

When applicable, and depending upon the specific substituents being 
present, the pteridine derivatives having the general formula (I) may be in the 
form of a pharmaceutically acceptable salt. The latter indude any 
therapeutically active non-toxic addition salt which compounds having the 

25 general formula (I) are able to form with a salt-forming agent. Such addition 
salts may conveniently be obtained by treating the pteridine derivatives of the 
invention with an appropriate salt-forming add or base. For instance, pteridine 
derivatives having basic properties may be converted into the corresponding 
therapeu-tically active, non-toxic add addition salt form by treating the free 

30 base form with a suitable amount of an appropiate add following conventional 
procedures. Examples of such appropnate salt-forming acids indude, for 
instance, inorganic adds resulting in forming salts such as but not limited to 
hydrohalides (e.g. hydrochloride and hydrobromide), sulfate, nitrate, 



28 



phosphate, diphosphate, carbonate, bicarbonate, and the like; and organic 
monocarboxylic or dicarboxylic acids resulting in forming salts such as, for 
example, acetate, propanoate, hydroxyacetate, 2-hydroxypropanoate, 2- 
oxopropanoate, lactate, pyruvate, oxalate, malonate, succinate, maleate, 
5 fumarate, malate, tartrate, citrate, methanesulfonate, ethanesulfbnate, 
benzoate. 2-hydroxybenzoate, 4-amino-2-hydroxybenzoate, benzenesulfo- 
nate, p-toluenesulfbnate, salicylate, p-aminosalicylate, pamoate, bitartrate. 
camphorsulfonate, edetate, 1,2-ethanedisulfonate, fumarate. glucoheptonate, 
gluconate, glutamate, hexylresorcinate, hydroxynaphtoate, hydroxyethane- 

10 sulfonate, mandelate, methylsulfate, pantothenate, stearate, as well as salts 
derived from ethanedioic, propanedioic, butanedioic, (Z)-2-butenedioic, (E)2- 
butenedioic, 2-hydroxybutanedioic, 2,3-dihydroxy-butanedioic, 2-hydroxy- 
1,2,3-propanetricarboxylic and cyclohexanesulfamic acids and the like. 

Pteridine derivatives having acidic properties may be converted in a 

15 similar manner into the corresponding therapeutically active, non-toxic base 
addition salt form. Examples of appropriate salt-forming bases include, for 
instance, inorganic bases like metallic hydroxides such as but not limited to 
those of alkali and alkaline-earth metals like calcium, lithium, magnesium, 
potassium and sodium, or zinc, resulting in the corresponding metal salt; 

20 organic bases such as but not limited to ammonia, alkylamines, benzathine, 
hydrabamine, arginine, lysine, N.N'-dibenzylethylenediamine, chloroprocaine, 
choline, diethanolamine, ethylene-diamine, N-methylglucamine, procaine and 
the like. 

Reaction conditions for treating the pteridine derivatives (I) of this 
25 invention with an appropriate salWorming acid or base are similar to standard 
conditions involving the same acid or base but different organic compounds 
with basic or acidic properties, respectively. Preferably, in view of its use in a 
pharmaceutical composition or in the manufacture of medicament for treating 
specific diseases, the pharmaceutically acceptable salt will be designed, i.e. 
30 the salt-forming acid or base will be selected so as to impart greater water- 
solubility, lower toxicity, greater stability and/or slower dissolution rate to the 
pteridine derivative of this invention. 



29 



The present invention provides the use of the above-described 
pteridine deivatives as biologically-active ingredients, i.e. active principles, 
especially as a medicine or a diagnostic agent or for the manufacture of a 
medicament or a diagnostic kit for the prevention or treatment of a TNF-a- 
related disorder in a mammal. The class of such disorders include the following: 

- septic or endotoxic shock or sepsis, especially in patients with a serum 
level of interleukin-6 above 1 ,000 pg/ml at start of treatment; 

- vascular TNF-a- mediated diseases such as, but not limited to, 
disseminated intravascular coagulation and Kawasaki's pathology; 

- pathologies and conditions associated with and/or induced by abnormal 
levels of TNF-a (herein defined as exceeding by at least 10 % and at 
most 500% the TNF-a level present in a normal healthy subject) 
occurring in a systemic, localized or particular tissue type or location in 
the body of the mammal; such tissue types include, but are not limited 
to, blood, lymph, liver, kidney, spleen, heart muscle or blood vessels, 
brain or spinal cord white matter or grey matter, cartilage, ligaments, 
tendons, lung, pancreas, ovary, testes and prostate. Abnormal TNF 
levels can also be localized to specific regions or cells in the body, such 
as joints, nerve blood vessel junctions and bones Such pathologies 
include alcohol-induced hepatitis, neurodegenerative diseases such as 
extrapyramidal and cerebellar disorders including lesions of the 
corticospinal system; disorders of the basal ganglia; hyperkinetic 
movement disorders such as chorea; drug-induced movement 
disorders; hypokinetic movement disorders, such as Parkinson's 
disease, spinocerebellar degenerations such as spinal ataxia, multiple 
systems degenerations (including Dejerine-Klumpke syndrome) and 
systemic disorders (including Refsum's disease, abetalipoprotemia, 
ataxia and telangiectasia); disorders of the motor unit, such as 
neurogenic muscular atrophies (anterior horn cell degeneration, such 
as amyotrophic lateral sclerosis, infantile spinal muscular atrophy and 
juvenile spinal muscular atrophy); Alzheimer's disease; Wernicke- 
Korsakoff syndrome; Creutzfeldt-Jakob disease; Hallerrorden-Spatz 
disease; and primary or secondary myelodysplasia syndromes; 



30 



- toxic effects of TNF-a and/or anti-cancer chemotherapeutic agents, 
especially side effects associated with TNF generation during neoplastic 
therapy, for instance following use of cisplatin; 

- injuries after irradiation of a tissue of a mammal by radio-elements, such 
5 as but not limited to radiation-induced graft-versus^host disease; and 

- cachexia and similar chronic wasting diseases, whether associated with 
cancer or with other chronic diseases such as malabsortive disorders, 
excessive physical stress, eating disorders and AIDS. 

The medicament of this invention may be for prophylactic use, i.e. where 
10 circumstances are such that an elevation in the TNF level might be expected or 
alternatively, may be for use in reducing the TNF level after it has reached an 
undesirably high level or as the TNF level is rising. 

The medicament according to this invention may be administered orally 
or in any other suitable fashion. Oral administration is preferred and the 

15 preparation may have the form of a tablet, aqueous dispersion, dispersable 
powder or granule, emulsion, hard or soft capsule, syrup, elixir or gel. The 
dosing forms may be prepared using any method known in the art for 
manufacturing these pharmaceutical compositions and may comprise as 
additives sweeteners, flavoring agents, coloring agents, preservatives and the 

20 like. Carrier materials and excipients are detailed hereinbelow and may 
include, inter alia, calcium carbonate, sodium carbonate, lactose, calcium 
phosphate or sodium phosphate; granulating and disintegrating agents, 
binding agents and the like. The pharmaceutical composition or combined 
preparation of this invention may be included in a gelatin capsule mixed with 

25 any inert solid diluent or carrier material, or has the form of a soft gelatin 
capsule, in which the ingredient is mixed with a water or oil medium. Aqueous 
dispersions may comprise the biologically active composition or combined 
preparation in combination with a suspending agent, dispersing agent or 
wetting agent Oil dispersions may comprise suspending agents such as a 

30 vegetable oil. Rectal administration is also applicable, for instance in the form 
of suppositories or gels. Injection (e.g. intramuscularly or intraperiteneously) is 
also applicable as a mode of administration, for instance in the form of 



31 

injectable solutions or dispersions, depending upon the disorder to be treated 
and the condition of the patient 

Usually the medicament of the invention is in the form of a combination 
of the pteridine derivative active principle and one or more pharmaceutically 
5 acceptable carriers or excipients 

The term " pharmaceutically acceptable carrier or excipient a as used 
herein refers to any material or substance with which the active principle, i.e 
the pteridine derivative having the general formula (I) may be formulated in 
order to facilitate its application or dissemination to the locus to be treated, for 

10 instance by dissolving, dispersing or diffusing the said composition, aid/or to 
facilitate its storage, transport or handling without impairing its effectiveness. 
The pharmaceutically acceptable carrier may be a solid or a liquid or a gas 
which has been compressed to form a liquid, i.e. the compositions of this 
invention can suitably be used as concentrates, emulsions, solutions, 

1 5 granulates, dusts, sprays, aerosols, pellets or powders. 

Suitable pharmaceutical earners for use in the said pharmaceutical 
compositions and their formulation are well known to those skilled in the art. 
There is no particular restriction to their selection within the present invention 
although, due to the usually low or very low water-solubility of the pteridine 

20 derivatives of this invention, special attention will be paid to the selection of 
suitable carrier combinations that can assist in properly formulating them in 
view of the expected time release profile. Suitable pharmaceutical earners 
include additives such as wetting agents, dispersing agents, stickers, 
adhesives, emulsifying or surface-active agents, thickening agents, 

25 complexing agents, gelling agents, solvents, coatings, antibacterial and 
antifungal agents (for example phenol, sorbic acid, chlorobutanol), isotonic 
agents (such as sugars or sodium chloride) and the like, provided the same 
are consistent with pharmaceutical practice, i.e. carriers and additives which 
do not create permanent damage to mammals. The pharmaceutical 

30 compositions of the present invention may be prepared in any known manner, 
for instance by homogeneously mixing, dissolving, spray-drying, coating 
and/or grinding the active ingredients, in a one-step or a multi-steps 



32 

procedure, with the selected carrier material and, where appropriate, the other 
additives such as surface-active agents, may also be prepared by 
micronisation, for instance in view to obtain them in the form of microspheres 
usually having a diameter of about 1 to 10 pm, namely for the manufacture of 
5 microcapsules for controlled or sustained release of the biologically active 
mgredient(s). 

Suitable surface-active agents to be used in the pharmaceutical 
compositions of the present invention are non-ionic, cationic and/or anionic 
materials having good emulsifying, dispersing and/or wetting properties. 

10 Suitable anionic surfactants include both water-soluble soaps and water- 
soluble synthetic surface-active agents. Suitable soaps are alkaline or 
alkaline-earth metal salts, unsubstituted or substituted ammonium salts of 
higher fatty adds (C10-C22), e 9 toe sodium or potassium salts of oleic or 
stearic acid, or of natural fatty acid mixtures obtainable form coconut oil or 

15 tallow oil. Synthetic surfactants include sodium or calcium salts of polyacrylic 
acids; fatty sulphonates and sulphates; sulphonated benzimidazole derivatives 
and alkylarylsulphonates. Fatty sulphonates or sulphates are usually in the 
form of alkaline or alkaline-earth metal salts, unsubstituted ammonium salts or 
ammonium salts substituted with an alkyl or acyl radical having from 8 to 22 

20 carbon atoms, eg. the sodium or calcium salt of lignosulphonic acid or 
dodecylsulphonic acid or a mixture of fatty alcohol sulphates obtained from 
natural fatty acids, alkaline or alkaline-earth metal salts of sulphuric or 
sulphonic acid esters (such as sodium lauryl sulphate) and sulphonic acids of 
fatty alcohol/ethylene oxide adducts. Suitable sulphonated benzimidazole 

25 derivatives preferably contain 8 to 22 carbon atoms. Examples of 
alkylarylsulphonates are the sodium, calcium or alcanolamine salts of 
dodecylbenzene sulphonic acid or dibutyl-naphtalenesulphonic acid or a 
naphtalene-sulphonic acid/formaldehyde condensation product. Also suitable 
are the corresponding phosphates, e.g. salts of phosphoric acid ester and an 

30 adduct of p-nonylphenol with ethylene and/or propylene oxide, or 
phospholipids. Suitable phospholipids for this purpose are the natural 
(originating from animal or plant cells) or synthetic phospholipids of the 
cephalm or lecithin type such as e.a. Dhosohatidylethanolamine. 



33 

phosphatidylserine, phosphatidylglycerine, lysoledlhin, cardiolipin, dioctanyl- 
phosphatidylcholine, dipalmitoylphoshatidylcholine and their mixtures. 

Suitable non-ionic surfactants include polyethoxylated and 
polypropoxylated derivatives of alkylphenols, fatty alcohols, fatty acids, 
5 aliphatic amines or amides containing at least 12 carbon atoms in the 
molecule, alkylarenesulphonates and dialkylsulphosuccinates, such as 
polyglycol ether derivatives of aliphatic and cycloaliphatic alcohols, saturated 
and unsaturated fatty acids and alkylphenols, said derivatives preferably 
containing 3 to 10 glycol ether groups and 8 to 20 carbon atoms in the 

1 0 (aliphatic) hydrocarbon moiety and 6 to 1 8 carbon atoms in the alkyl moiety of 
the alkylphenol. Further suitable non-ionic surfactants are water-soluble 
adducts of polyethylene oxide with poylypropylene glycol, 
ethylenediaminopolypropylene glycol containing 1 to 10 carbon atoms in the 
alkyl chain, which adducts contain 20 to 250 ethyleneglycol ether groups 

15 and/or 10 to 100 propyleneglycol ether groups. Such compounds usually 
contain from 1 to 5 ethyleneglycol units per propyleneglycol unit. 
Representative examples of non-ionic surfactants are nonylphenol- 
polyethoxyethanol, castor oil polyglycolic ethers, polypropylene/ polyethylene 
oxide adducts, tributylphenoxypolyethoxyethanol, polyethyleneglycol and 

20 octylphenoxypolyethoxyethanol. Fatty acid esters of polyethylene sorbitan 
(such as polyoxyethylene sorbitan trioleate), glycerol, sorbitan, sucrose and 
pentaerythritol are also suitable non-ionic surfactants. 

Suitable cationic surfactants include quaternary ammonium salts, 
preferably halides, having 4 hydrocarbon radicals optionally substituted with 
25 halo, phenyl, substituted phenyl or hydroxy, for instance quaternary 
ammonium salts containing as N-substituent at least one Ce-C^ alkyl radical 
(e g cetyl, lauryl, palmityl, myristyl, oleyl and the like) and, as further 
substituents, unsubstituted or halogenated lower alkyl, benzyl and/or hydroxy- 
lower alkyl radicals 

JO A more detailed description of surface-active agents suitable for this 

purpose may be found for instance in "McCutcheon's Detergents and 
Emulsifiers Annual" (MC Publishing Crop., Ridgewood. New Jersey, 1981), 



34 



"Tensid-Taschenbuch", 2 nd ed. (Hanser Verlag, Vienna, 1981) and 
"Encyclopaedia of Surfactants (Chemical Publishing Co., New York, 1981). 

Structure-forming, thickening or gel-forming agents may be included into 
the pharmaceutical compositions of the invention. Suitable such agents are in 
5 particular highly dispersed silicic acid, such as the product commercially 
available under the trade name Aerosil; bentonites; tetraalkyl ammonium salts 
of montmorillonites (e.g., products commercially available under the trade 
name Bentone), wherein each of the alkyl groups may contain from 1 to 20 
carbon atoms; cetostearyl alcohol and modified castor oil products (e.g. the 

1 0 product commercially available under the trade name Antisettle). 

Gelling agents which may be included into the pharmaceutical 
compositions of the present invention include, but are not limited to, cellulose 
derivatives such as carboxymethylcellulose, cellulose acetate and the like; 
natural gums such as arabic gum, xanthum gum, tragacanth gum, guar gum 

15 and the like; gelatin; silicon dioxide; synthetic polymers such as carbomers, 
and mixtures thereof. Gelatin and modified celluloses represent a preferred 
class of gelling agents. 

Other optional excipients which may be included in the pharmaceutical 
compositions of the present invention include additives such as magnesium 

20 oxide; azo dyes; organic and inorganic pigments such as titanium dioxide; UV- 
absorbers; stabilisers; odor masking agents; viscosity enhancers; antioxidants 
such as, for example, ascorbyl palmitate, sodium bisulfite, sodium 
metabisulfite and the like, and mixtures thereof; preservatives such as, for 
example, potassium sorbate, sodium benzoate, sorbic acid, propyl gallate, 

25 benzylalcohol, methyl paraben, propyl paraben and the like; sequestering 
agents such as ethylene-diamine tetraacetic acid; flavoring agents such as 
natural vanillin; buffers such as citric acid and acetic acid; extenders or bulking 
agents such as silicates, diatomaceous earth, magnesium oxide or aluminum 
oxide; densification agents such as magnesium salts; and mixtures thereof. 

30 Additional ingredients may be included in order to control the duration of 
action of the biologically-active ingredient in the compositions of the invention. 
Control release compositions may thus be achieved by selecting appropriate 



35 

polymer carriers such as for example polyesters, polyamino-acids, polyvinyl- 
pyrrolidone, ethylene-vinyl acetate copolymers, methylcellulose, 
carboxymethylcellulose, protamine sulfate and the like. The rate of drug 
release and duration of action may also be controlled by incorporating the 
5 active ingredient into particles, e.g. microcapsules, of a polymeric substance 
such as hydrogels, polylactic acid, hydroxymethyl-cellutose, polymethyl 
methacrylate and the other above-described polymers. Such methods include 
colloid drug delivery systems like liposomes, microspheres, microemulsions, 
nanoparticles, nanocapsules and so on Depending on the route of 
0 administration, the pharmaceutical composition of the invention may also 
require protective coatings. 

Pharmaceutical forms suitable for injectable use include sterile aqueous 
solutions or dispersions and sterile powders for the extemporaneous 
preparation thereof. Typical carriers for this purpose therefore include 
5 biocompatible aqueous buffers, ethanol, glycerol, propylene glycol, 
polyethylene glycol, complexing agents such as cyclodextnns and the like, 
and mixtures thereof. 

The present invention further relates to a method for preventing or treating 
a TNF-a-related disease, such as above defined, in a subject or patient by 
> administering to the patient in need thereof an effective amount of a pteridine 
derivative having the general formula (I). The effective amount is usually in the 
range of 0.01 mg to 20 mg, preferably 0.1 mg to 5 mg, per day per kg 
bodyweight for humans. Depending upon the pathologic condition to be 
treated and the patient's condition, the said effective amount may be divided 
into several sub-units per day or may be administered at more than one day 
intervals. The patient to be treated may be any warm-blooded animal such as 
a mammal, preferably a human being, suffenng from said TNF-a-related 
disease. 

The following examples are intended to illustrate several embodiments of 
the present invention, including the preparation of the pteridine derivatives, 
without limiting its scope in any way. 

Example 1 - preparatio n of 2-aminQ^4-n-oentvlow-6-stvrvlDteridine 



10 



30 



36 

A mixture of 1.5 g (5.6 mmoles) 2^mino^-chloro-4-n-pentyloxypteridine 
(e.g. available following the procedure disclosed by Mohr et al. in Hetv. Chem. 
Acta (1992) 75:2317), palladium acetate (63 mg, 0.28 mmoles), tri-o- 
tolylphosphane (682 mg, 2.24 mmoles), cuprous iodide (53 mg, 0.28 mmoles), 
styrene (1,3 ml., 11.3 mmoles) and triethylamine 3.1 ml, 22 mmoles) was 
stirred in dry acetonitrile (50 ml) under reflux for 90 hours. It was evaporated 
and the residue purified by silica gel column chromatography with chloroform. 
The product fraction was evaporated to give 1.37 g (yield: 72%) of an orange 
powder exhibiting, after recrystallization from a EtOAc/hexane mixture, a 
melting point (m p.) range of 127-128 °C. 

Example 2 - preparation of 2^amino-6-(l 2-dibromoDhfinBthvlM-n-Dentvlnyy- 
pteridine . 

To a solution of the denvative of example 1 (1.0 g, 2.94 mmoles) in 
15 chloroform (50 ml) was added a 2 M bromine solution in chloroform (2.2 ml., 
4.4 mmoles) and then the mixture was stirred at room temperature for 7 hours. 
It was diluted with chloroform (50 ml), washed with a saturated aqueous 
NazSOa solution (100 ml) and dried over sodium sulfate. After evaporation of 
the solvents, the residue was treated with toluene, filtered, washed with ether 
and dried in a vacuum desiccator to give 0.84 g (yield: 57%) of a yellow 
powder. 



Example 3 - preparation of 2-amino-4 7-dimethoxv-6-stvry|rteridine 

A suspension of the derivative of example 2 (0.3 g, 0.6 mmoles) is 
methanol (10 ml) was treated with 1 M methanol sodium methoxide (3 ml, 3 
mmoles) and then refluxed for 4 hours. It was diluted with chloroform (100 ml), 
washed with a saturated aqueous ammonium chloride solution and water and 
then the solution was dried over sodium sulfate. The filtrate was evaporated 
and the residue was purified by silica gel column chromatography while using 
chloroform as the eluent. The product fraction was evaporated to give 50 mg 
(yield: 26%) of a yellow powder with a melting point range of 1 97-1 98 °C. 



20 



25 



37 

Example 4 - preparation of Q 4 -methvl-biop terin f2-amino-4-methoxv-6-(1 ,2- 
dihvdroxvpropvn pteridine) 

To a solution of N 2 , 1', 2'-0-triacetyl-biopterin (1,0 g, 2.75 mmoles), 
triphenyl-phosphane (12,08 g, 4.13 mmoles) and methanol (0.15 ml, 3.7 
mmoles) in dry dioxane (30 ml) was added diisopropyl azodicarboxylate (0.81 
g, 4.1 mmoles). After stirring for 1.5 hour at room temperature, the mixture 
was evaporated to dryness. The residue was purified by silica gel column 
chromatography while using an ethylacetate/CHCI 3 (1:4) mixture as the 
eluent. The product fraction was evaporated and dried in vacuum to give 0.4 g 
(yield: 38%) of N 2 ,1',2 , -0-triacetyl-0 4 -methylbiopterin. Deacetylatton of this 
reaction product (0.28 g, 074 mmole) was done by stirring it in absolute 
methanol (20 ml) and triethylamine (4 ml) for 24 hours. Evaporation to 
dryness, treatment of the residue with ether, filtration and drying gave 0.172 g 
(yield: 83%) of 0 4 -methyl-biopterin with a melting point range of 160-161 °C. 

Example 5 - preparation of 2^amino-4-hvdroxvlami nty6-phenvlpteridine. 
A suspension of 2,5,6-tnamino-4-methoxypyrimidine dihydrochloride (1 g, 4 
mmoles) in methanol (40 ml) was heated to boiling and then a solution of 
phenylglyoxalmonoxime (1 g, 6.6 mmoles) in methanol (10 ml) was added 
dropwise. A clear solution is obtained from which on reflux for 2 hours a 
precipitate was separated out. The solid (hydrochloride salt) was filtered off, 
suspended in water (30 ml) and then neutralized to pH 8 by concentrated 
ammonia. The resulting precipitate was collected, washed with water and 
ethanol and dried at 100 °C to give 0.84 g of a yellow powder (yield: 82%). 

Examples 6 to 53 - synthesis of 2-fl mino4-dialkvlamino-6-arvlPteridines. 2- 
amino-4-difarvlalkvhamino-6-arvlpteridine s 2-amin(M-alkvlamino-6-arylpten- 
dines. 2-amino-4-(N-containina heter ocyclic aminol-6-arvlPteridines and 2- 
amino-4^lkoxv-6-arvlpteridines 

The procedure for the synthesis of the following 2-amino-4-dialkylamino-6- 
arylpteridines, 2-amino-4-dialkylamino-6-arylpteridines, 2-amino-4-alkylamino- 
6-arylpteridines, 2-amino-4-(N-containing heterocyclic amino)-6-arylpteridines 
and 2-amino-4-alkoxy-6-arylpteridines proceeds in three steps: 



38 

) a solution of 2 l 6^iamino-4^loro^i>K^lorophenylazopyrimidine (a 
compound known from British Patent No. 677,342) (5.0 g, 16.6 mmoles) in 
DMF (50ml) and 0.12 mole of the appropriate reactant, being selected from 
the group consisting of secondary alkylamines and arylalkylamines (e.g. 
dimethyl-amme in ethanol (50%), diethylamine, di-n-propylamine or 
dibenzylamine), primary amines (e.g. an adamantanamine), heterocyclic 
amines (e.g. morpholine, piperidine, pyrrolidine, piperazine or N-methyl- 
piperazine) and alcaline metal alkoxides (e.g. sodium ethoxide or sodium 
isopropoxide), were heated in an oil bath at 70°C for 5 hours. Then water 
(50 ml) was added, cooled and the yellow precipitate collected, washed 
with water and dried. Recrystallization from ethanol or a DMF/water 
mixture provided the relevant 2,6-diamino4-dialkylamino-5-p-chloro- 
phenylazopyrimidine, 2 f 6^iamino4-di(arylalkyl)amino-5-p-chlorophenyl- 
azopyrimidine, 2 f 6^iamino^^lkyl-amino-5-p-chlorophenylazopyrimidine, 
2,6-diamino-4-(N-containing hetero-cyclic amino)-5-p-chlorophenylazo- 
pyrimidine or 2,6^iamino-4-alkoxy-5-p-chlorophenylazopyrimidine with a 
yield ranging from 55 to 90% 

a suspension of the pyrimidine compound (3.28 g, 10 mmoles) resulting 
from step (a) in methanol (70 ml) and concentrated ammonia (10 ml) was 
reduced in a shaking apparatus under a hydrogen atmosphere in the 
presence of a Raney nickel catalyst (3.5 g) for 2 days. The catalyst was 
filtered off under argon atmosphere and then the filtrate evaporated in 
vacuo to dryness. The residue was treated with ether to remove p- 
chloroanilme, filtered and then the solid stirred in methanolic HCI (10%, 50 
ml) overnight. The dihydrochloride salt (obtained with a yield ranging from 
85 to 90%) of the relevant 2,5 l 6-triamino-4-dialkylaminopyrimidine, 2,5,6- 
triamino-4-alkoxypyrimidine, 2,5,6-tnamino-4-di(arylalkyl)aminopyrimidine, 
2,5,6-triamino-4-alkylaminopyrimidine or 2,5,6-triamino-4-(N-containing 
heterocyclic amino) pyrimidine, was collected and dried in a vacuum 
desiccator over KOH 

to a boiling solution of the 2,5.6-triamino-4-substituted pyrimidine 
dihydrochloride salt (5 mmoles) from step (b) in methanol (20 ml) was 
added a solution of the relevant arylglyoxalmonoxime (7 5 mmoles) in 



39 



methanol (10 ml) dropwise and then the mixture was heated under reflux 
for 3 hours. After cooling, the suspension or solution was made alkaline by 
means of concentrated ammonia up to pH 9 and the resulting precipitate 
was filtered off, washed with water and dried. Recrystallization was done 
from ethanol and a DMF/water mixture, respectively, such as to provide a 
yellow solid with a yield ranging from 50 to 85%. 

The following compounds were prepared according to the above general 

procedure: 

2-amino-4-dimethylamino-6»phenylpteridine (example 6); 
2-amino-4-dimethylamino^-(4-tolyl) pteridine (example 7); 
2-amino-4-dimethylamino-6-<4-methoxyphenyl)pteridine (example 8); 
2-amino-4-diethylamino-6-phenylpteridine (example 9); 
2-amino-4-diethylamino-6-(4-chlorophenyl)pteridine (example 10); 
2-amino-4-diethylamino^-(4-methoxyphenyl)pteridine (example 11); 
2^mino-4<liethylamino^3,4^imethoxyphenyl)pteridine (example 12); 
2-amino-4-dibenzylamino-S-phenylpteridine (example 13); 
2^mino^ibenzylamino-6-(4-chlorophenyl)pteridine (example 14); 
2^mino-4HJibenzylamino^4-methoxyphenyl)pteridine (example 15); 
2^mino^ibenzylamino-6-(3,4-dimethoxyphenyl)pteridine (example 16); 
2^mincHl-dipropylamino-6-phenylpteridine(example 1 7); 
2^mino^ipropylamino-6-(4-chlorophenyl)pteridine (example 18); 
2-amirx)^ipropylamino-6-(4-methoxyphenyl)pteridine (example 19); 
2^mino-4<lipropylamino^^3 f 4^imethoxyphenyl)pteridine (example 20); 
2^mino-4-morpholino^-phenylpteridine (example 21 ); 
2^mino-4^onpholino-6^4-chlorophenyl)pteridine (example 22), 
2-amino-4-morpholino-6-(4-methoxyphenyl)pteridine (example 23); 
2-amino«4-morpholmo-6-(3,4-dimethoxyphenyl)pteridine (example 24); 
2-amino-4-piperidino-6^)henylpteridine (example 25); 
2-amino-4-piperidino^-{4-chlorophenyl)pteridine (example 26); 
2-amino-4-pipendino-6-(4-methoxyphenyl)pteridine (example 27); 
2-amino-4-pipendino-6-(3,4-dimethoxyphenyl)pteridine (example 28); 
2-amino-4-N-methylpiperazino-6-phenylpteridine (example 29); 
2-amino-4-N-methylpiperazino-6-(4-chlorophenyl)pteridine (example 30); 



40 



2-amino^N-methylpiperazino-6- (4-methoxyphenyl) pteridine (example 31); 
2-amino-4-methylpiperazino-6- (3, 4-dimethoxyphenyl) pteridine (example 32); 
2-amino-4-pyrrolidino-6- (4-methoxyphenyl) pteridine (example 33); 
2-amino-4-piperazino-6-phenylpteridine (example 34); 
2-amino-4-piperaano-6- (4-chlorophenyl) pteridine (example 35); 
2-amino-4-piperazino-6- (4-methoxyphenyl) pteridine (example 36); 
2-amino-4-piperazino-6- (3, 4-dimethoxyphenyl) pteridine (example 37); 
2^mino-4-dibenzylamino-6-{3 l 4,5-trimethoxyphenyl) ptendine (example 38); 
2-amino-4-morpholino-6- (3, 4, 5-trimethoxyphenyl) pteridine (example 39); 
2-amino-4-(3-adamantylamino)-6-{3 l 4 l 5-trimethoxyphenyl) pteridine (example 
40); 

2-amino^-(3-adamantylamino)-6-naphtylpteridine (example 41 ); 
2-amino-4-(4-adamantylamino)-6-(3 l 4 1 5-trimethoxyphenyl) pteridine (example 
42); 

2-amino-4-(4-adamantylam»no)-6-naphtylpteridine (example 43); 

2^mino-4-morpholino^3,4-fo(^^ 

(example 44); 

2-amino-4-dimethylamino^3,4-fonrnylidene-3,4-dihydroxyphenyl)pteridine 
(example 45); 

2-amino-4-pyrroltdino-6-(3, 4-dimethoxyphenyl) pteridine (example 46); 
2-amino-4-dimethylamino-6-{3, 4-dimethoxyphenyl) ptendine (example 47); 
2-amino-4-dimethylamino-6-methylpteridme (example 48); 
2-amino-4-ethoxy-6-phenylpteridine (example 49); 
2-amino-4-propylamino^-phenylpteridine (example 50); 
2-amino-4-propylamino-6-(3 1 4-dimethoxyphenyl) pteridine (example 51 ); 
2-acetamido4HSopropoxy-6-(3 > 4-dimethoxyphenyl) pteridine (example 52); 
and 

2~amino-4-ethoxy-6-(3,4-dimethoxyphenyl)pteridine (example 53); 

Example 54 - synthesis of 2.6^iamino-4-ethoxv-pyrimidine 

To a solution of sodium (1 .05 g) in ethanol (50 ml) was added 4-chloro- 
2,6-diaminopyrimidme (6 g t 41 4 mmoles). The resulting solution was heated 
in a reactor for 6 hours at 160 °C. The reaction mixture was cooled down and 



41 

the precipitated sodium chloride was filtered off. The filtrate was concentrated 
and precipitated from ethanol (two times), affording the pure title compound as 
a white solid (4.53 g, 72% yield). The spectral data are identical to those 
described e.g. by W. Pfleiderer et al. in Chem. Ber. (1961 ) 94, 12 

Example 55 - synthesis of 2.6-diamino-4-isoDropoxv-ovrimidine 

The same procedure as in example 54 was followed using isopropanol 
instead of ethanol. The filtrate was pure enough for further reaction without 
purification The spectral data are identical to those described e.g. by W. 
Pfleiderer etal. in Chem. Ber. (1961) 94, 12. 

Example 5 6 - synthesis of 5-nitros6-2.6-diamino-4-ethoxv-ovrimidine 

To a solution of the compound of example 54 (6.13 g, 39.8 mmoles) in 
20 % aqueous acetic acid (57 ml) was added dropwise a solution of NaN0 2 
(3.29 g) in water (13 ml) at 80 °C. A pink precipitate was formed which was 
stirred at 80°C for an additional 2 hours. The reaction mixture was cooled 
down in the refrigerator overnight and the resulting precipitate was filtered off, 
yielding the title compound as a pink powder (4 98 g, yield 68 %). Spectral 
data are identical with those described e.g. by W. Pfleiderer et al. in Chem. 
Ber (1961) 94, 12. 

Example 57 - synthesis of 5-nitroso-2.6-diamino-4-isopropoxv-pvrimidine 

The same procedure was followed as in example 56 but starting from 
the compound of example 55. The product has identical spectral data to those 
described by W. Pfleiderer et al. (cited supra). 

Example 58 - synthesis of 2.5.6-triamino-4-ethoxv-pvrimidine 

To a suspension of the compound of example 56 (7.12 g, 38.9 mmoles) 
in water (150 ml) at 60 °C was added sodium dithionite (46.7 mmol, 8.12 g). 
Additional sodium dithionite was added till the pink colour completely 
disappeared and a yellow solution was formed. The solution was stirred at 60 
°C for another 4 hours. Water was evaporated and the resulting residue was 
precipitated from a small amount of water, providing the title compound as a 



42 

yellow powder (4.02 g, yield 61 %) Spectral data are identical with literature 
data (W. Pfleiderer et al cited supra). 

Example 59 - synthesis of 2,5.6-triamino^sopropoxvHPvrimidine 
5 The procedure of example 58 was followed, however using the 

compound of example 57 as the starting material. The spectral data of the 
product obtained are identical with the literature data (W. Pfleiderer et al. 
cited supra). 

Example 60 - synthesis of 2-amino-4-ethoxv-pteridin 

10 To a solution of 2 l 5,64riamino4-ethoxy-pyrimidine (10.54 g, 62.37 

mmoles) in ethanol (160 ml) was added glyoxal (40 % solution in water, 2.7 
ml, 18.6 mmoles). The reaction mixture was refluxed for 4 hours. Some 
insoluble material was filtered off. The filtrate was concentrated in vacuo and 
the residue purified by flash chromatography (silica, using a CH3OH/CH2CI2 

15 mixture (5:95) as the eluent), providing the pure title compound (7.34 g, yield: 
62 %) The spectral data of the product are identical with the literature data 
(W. Pfleiderer et al. cited supra) 

Example 61 - synthesis of 2-amino^sopropoxv-pteridin 
20 The procedure of example 60 was repeated, however using 

isopropanol as the solvent instead of ethanol. The spectral data of the product 
obtained are identical with the literature data (W. Pfleiderer et al. cited supra). 

Example 62 - synthesis of 2-amino-4-ethoxvpteridine-N 8 -oxide 
25 To a cooled (0 °C) solution of the compound of example 60 (2.47 g, 

12.9 mmoles) in trifluoroacetic acid (53 ml) was added dropwise 2.53 ml of a 
35 % aqueous H2O2 solution. The reaction mixture was kept at 4 °C for two 
days in the refrigerator, whereby another 1.25 ml of the same H2O2 solution 
was added after 1 day. The solution was concentrated in vacuo. The residue 
30 was suspended in water and neutralized by the addition of a concentrated 
ammonia solution. Evaporation of the solvent in vacuo and purification of the 
residue by flash chromatography (silica, using a CH3OH/CH2CI2 mixture (6:94) 
as the eluent) provided the title compound as a yellow powder (861 mg, yield: 



43 

32 %). Mass spectrum data are as follows: m/z (%) 230 ([M+Naf , 30), 208 
([M+H]*, 100), 180 [(M+H-ethene)*, 10]. 

Example 63 - synthesis of 2-arnino-4-isooropoxvpteridine-N 6 -oxide 

The procedure as described in example 62 was followed, however 
using the compound of example 61 as the starting material. Mass spectrum 
data are as follows: m/z (%): 222 ([M+Hf, 100), 180 ([M+H-propenef , 60) 

Example 64 - synthesis of 2^amino-6-chlon>4-ethoxvDteridine 

A suspension of the compound of example 62 (460 mg, 2.22 mmoles) 
in acetyl chloride (5.5 ml) was stirred at -40 °C. Trifluoroacetic acid (1.69 ml) 
was then added dropwise. The resulting solution was slowly warmed up to 0 
°C and stirred for an additional 4 hours at 0 °C. Reaction was carefully 
quenched with ice, followed by neutralization with a concentrated ammonia 
solution (pH = 8). The aqueous phase was extracted with CH 2 CI 2 (five times). 
The combined organic layers were concentrated in vacuo and the residue was 
purified by flash chromatography (silica, using a CH 3 OH/CH 2 CI 2 mixture (1:99) 
as the eluent), thus providing the title compound as a yellow powder (360 mg, 
yield: 72 %). This compound was further characterized as follows: 

- mass spectrum: mfc (%): 226 ([M+H]*, 1 00), 

- 1 H-NMR (200 MHz, DMSO-d 6 ): 5 1.42 (3 H, t), 4.52 (2 H, q), 7.42 (2 H, 
d) and 8.85 (1 H,s)ppm, 

- 13 C-NMR (50 MHz, DMSCW 6 ): 5 14.19, 63.58, 121.74, 140.22, 150.99, 
156.13, 161.98 and 165.97 ppm. 

Example 65 - synthesis of 2-aminch6-chloro-4HSopropoxvpteridine 

The procedure as described in example 64 was followed, however 
starting from the compound of example 63 The mass spectrum data of the 
resulting compound are as follows: m/z (%): 240 ([M+Hf, 55), 198 ([M+H- 
propene]*, 100). 



44 

Examples 66 to 83 - synthesis of 2-amino-6-arvl-4-ethoxvptendines and 2- 
amino-6-heteroarvl-4-ethoxvpteridines 

The general procedure used for preparing 2-amino-6-aryl-4-ethoxy- 
pteridines is as follows, to a degassed solution of the compound of example 
5 64 (50 mg, 0.22 mmole) in THF (5 ml) was added a degassed solution of 
sodium carbonate (5 ml of a 0.4 M solution in water), tetrakis(triphenyl- 
phosphine) palladium (0.013 mmole, 14 mg) and an arylboronic or (examples 
72 and 73) heteroarylboronic acid (0.22 mmole). The solution was refluxed for 
4 hours Solvents were concentrated in vacuo and the residue was purified by 

10 flash chromatography (silica) with an appropriate CH 3 OH/CH 2 CI 2 mixture (2:98 
or 3:97) as the eluent (except for the compound of example 82, which was 
eluted with an acetone/CH 2 CI 2 (7:3) mixture). This procedure provided, with a 
yield ranging from 16 % to 60 % depending upon the aryl or heteroaryl group 
(from the arylboronic or heteroarylboronic acid) introduced at the 6-position of 

15 the pteridine ring, the following pure final compounds which were 
characterized by their mass spectrum MS and optionally by their 'H-NMR (200 
MHz, DMSO-cfe) spectrum. 

- 2-amino-6-(p-methoxyphenylH-ethoxy-pteridine (example 66): MS 298 
([M+Hf, 100), 270 ([M+H-ethenef, 55); 

20 - 2^minc>^o^thoxyphenylH-ethoxy-pteridine (example 67): MS 298 
([M+Hf, 100), 270 ([M+H-ethenef, 30); 

- 2^mino-6-<m-methoxyphenylH-ethoxy-pteridine (example 68): MS 
298 <[M+Hf, 100), 270 ([M+H-ethenef, 35); 'H-NMR: 1.46 (3 H, t), 
3.85 (3 H. s). 4 58 (2 H, q), 7.06 (1 H, dd). 7.33 (2 H, br s), 7.46 (1 H, t), 

25 7.68(1 H,m) and 9 43(1 H,s)ppm; 

- 2-amino-6-<3,4-difluorophenyl)-4-ethoxy-pteridine (example 69): MS 
304 ([M+Hf ,100), 270 ([M+H-ethenef, 35); 'H-NMR: 1.45 (3 H, t), 4.57 
(2 H, q), 7.42 (2 H, br s), 7.60 (1 H, q), 7.98 (1 H, d), 8.16 (1 H, t) and 
9.42 (1 H, s) ppm; 

30 - 2-amino-6-(p-dimethylaminophenyl)-4-ethoxy-ptendine (example 70): 
MS 311 ([M+Hf ,100), 283 ([M+H-ethenef, 35); 

- 2-amino-6-(p-trifluoromethylphenyl)-4-ethoxy-pteridine (example 71): 
MS 336 ([M+Hf ,100), 308 ([M+H-ethenef, 50); 



45 



- 2-amino-6^2-thienyl)-4-ethoxy-pteridine (example 72): MS 274 ([M+H]*, 
100), 246 ([M+H-ethene]*, 40); 

- 2-amino-6-(3-thienyl)-4-ethoxy-ptericline (example 73): MS 274 ([M+H]*, 
100), 246 ([M+H-ethener, 45); 

- 2^mino-6-(3,4-dichlorophenyl)-4-«thoxy-pteridine (example 74): MS 
337 ([M+H]*,1O0); 'H-NMR: 1.46 (3 H, t), 4.59 (2 H, q), 7.42 (2 H, br s), 
7.81 (1H, d), 8.14 (1 H, dd), 8.37 (1 H, d) and 9.47 (1 H, s) ppm; 

- 2-amino-6-(p-cyanophenyl)-4-ethoxy-pteridine (example 75): MS 293 
({M+H]*. 100), 265 ([M+H-ethener, 65); 

- 2-amino-6-{p-ethoxyphenyl)-4-ethoxy-pteridine (example 76): MS 312 
([M+H]*, 100), 284 ([M+H-ethene] + , 70); 

- 2-amino-6-(p-fluorophenyl)-4-ethoxy-pteridine (example 77): MS 286 
([M+H]*, 100), 258 ([M+H-ethene)*, 45); 

- 2-amino-6^p-ethylphenyl)-4-ethoxy-pteridine (example 78): MS 296 
([M+H]*, 100), 268 ([M+H-ethene)*, 45); 

- 2-amino-6-(p-acetylphenyl)-4-ethoxy-pteridine (example 79): MS 310 
([M+H]*, 1 00), 282 ([M+H-ethene]*. 60); 

- 2-amino-6-(3-methyl-4-fluorophenyl)-4-ethoxy-pteridine (example 80): 
MS 300 ([M+H]*, 1 00), 272 ([M+H-ethene]*, 30); 

- 2-amino-6-(p-thiomethylphenyl)-4-ethoxy-pteridine (example 81): MS 
314 ([M+H]*, 100), 286 ([M+H-ethene]*, 35); 

- 2-amlno-6-(p-N,N-dimethylbenzamido)-4-ethoxy-pteridine (example 82) 
MS 338 ([M+H]*, 100), 311 ([M+H-ethene]*, 15), and 

- 2-amino-6-(3,4-dimethoxyphenyl)-4-ethoxy-ptendjne (example 83): MS 
328 ([M+H]*, 100), 300 ([M+H-ethene]*, 40). 

Examples 84 to 98 - synthesis of 2-amino-6-arvl-4-isopropoxypteridines and 2- 
amino-6-heteroan/l-4-isopropoxvpteridines 

The procedure as described in examples 66-83 was followed while 
using 2-amirK)-6-chloro-4-isopropoxypteridine as the starting material, except 
that longer reaction times were needed (refluxing overnight instead of 4 
hours). This procedure provided, with a yield ranging from 10 % to 70 % 
depending upon the aryl or heteroaryi group introduced at the 6-position of the 



46 



ptendine ring, the following pure final compounds which were characterized by 
their mass spectrum: 

- 2^mino^3^ethyl-4-methoxyphenyl)-4-isopropoxypteridine (example 
84): MS 326 ([M+HjMOO), 284 ([M+H-propene]*, 30); 

- 2^mino^3,4^1imethylphenyl)-4-isopropoxypteridine (example 85): 
MS 310 ([M+Hf.lOO), 268 ([M+H-propene]\ 60); 

- 2^minc^3^loro-44rifluoromemylphenyl)^soprt^xypteridine 
(example 86). MS 384 ([M+H]*, 20), 342 ([M+H-propene]*, 50); 

- 2^mino^^3-chloro-4-fluorophenyl)-4-isopropoxypteridine (example 

87) : MS 334 ([M+H]*, 20), 292 ([M+H-propene]*, 50); 

- 2-amino-6-(p-N,N-diethylbenzamido)-4Hsopropoxypteridine (example 

88) : MS 381 ([M+Hf, 100); 

- 2^minc^p-trffluoromethylphenyl)-4H'sopropoxypteridine (example 

89) : MS 350 ([M+H]*, 1 00), 308 ([M+H-propene]*, 30); 

- 2-amino-6-(3,4-difluorophenyl)-4Hsopropoxypteridine (example 90): MS 
31 8 ([M+H]*, 100), 276 ([M+H-propene]*, 50); 

- 2-amino^-(p-methoxyphenyl)-4-isopropqxypteridine (example 91): MS 
312 ([M+H]* ( 100), 270 ([M+H-propene]*, 50); 

- 2-amino^-(p-ethoxyphenyl)-4-isopropoxypteridine (example 92). MS 
326 ([M+H]*, 55), 284 ([M+H-propener. 1 00); 

- 2^mino-6-(p-dimethylben2amido)-4-isopropoxypteridine (example 93): 
MS 353 ([M+H]*, 75), 311 ([M+H-propener, 100); 

- 2-amino-6-(3-thienyl)-4-isofropoxypteridine (example 94): MS 288 
([M+H]*. 55), 246 ([M+H-propene]*. 100); 

- 2-amino-6-(p-cyanophenyl)-4-isopropoxypteridine (example 95): MS 
307 ([M+H]*, 40), 265 ([M+H-propene]*. 100); 

- 2-amino-6-(p-benzoic acid methyl ester)-4-isopropoxypteridine (exam- 
ple 96): MS 340 ([M+H]*, 75), 298 ([M+H-propene]*, 100); 

- 2-amino-6-(p-acetylphenyt)-4-isopropoxypteridine (example 97) MS 
324 ([M+H]*, 55), 282 ([M+H-propene]*, 1 00); and 

- 2-amino-6-(3,4-dimethoxyphenyl)-4-isopropoxypteridine (example 98): 
MS 342 ([M+Hr. 100), 300 ([M+H-propene]*, 60). 



47 

Example 99 - synthesis of 2.6-diamino-5^iitroso-4-hvdroxvpvnmidine 

To a solution of 2,6-diamino4-hydroxypyrimidine (12.9 g, 102.2 
mmoles) in 200 ml of a 10% acetic acid solution in water at 80 °C was added 
dropwise a solution of NaN0 2 (7.05 g, 102.2 mmoles) in 20 ml water. A pink 
5 preciprtate was formed, which was further stirred for 1 hour at 80 °C. The 
reaction mixture was cooled down in the refrigerator overnight. The precipitate 
was filtered off and dried over P 2 0 5> providing the title compound as a pink 
powder (15.43 g, yield: 97%). The spectral data are in accordance with 
literature data (Landauer et al. in J. Chem. Sac. (1953) 3721-3722). 

10 

Example 100 - synthesis of 2.5.64riamino-4-hydroxypvrimidine 

A suspension of the compound of example 99 (15 g, 96.7 mmoles) in 
an ammonium sulfide solution (20 % in water, 200 ml) was stirred overnight at 
50 °C The reaction mixture was cooled down in the refrigerator and the 
15 precipitate was filtered off, providing the title compound as a yellow powder 
(11.33 g, yield: 83 %). The spectral data are identical with literature data 
(Landauer et al. ated supra). 

Example 101 - synthesis of 2-amino-6-(3 t 4<iimethoxyphenyl)pterine 
20 To a boiling solution of the compound of example 100 (2.4 g, 17 

mmoles) in methanol (100 ml, with 0.9 N HCI) was added dropwise a solution 
of 3,4-dimethoxyphenylglyoxal mono-oxime (3.8 g, 18 mmoles) in methanol 
(100 ml). The reaction mixture was heated under reflux for 4 hours. The 
precipitate formed was filtered off, washed with water, then ethanol and 
25 diethyl ether, and dried over P2O5 under vacuum, providing the title compound 
as a yellow powder (4.33 g, yield: 85 %). This compound was further 
characterized by the following spectra: 

- ! H-NMR (500 MHz, TFA): 6 4.11 (3 H, s), 4.07 (3 H, s), 7.21 (1 H, d), 
7.78 (1 H, dd), 7.81 (1 H, d) and 9.32 (1 H, s) ppm; 
30 - 13 C-NMR (125 MHz, TFA): 6 56.39, 56,7, 111.94, 113.21, 123.22, 
127.41, 127.91, 145.92, 149.39, 150.46, 152 47, 153.15, 155.13 and 
161.59 ppm. 



48 



Example 102 - sy nthesis of 2-acetvlamino-6-(3.4-<jimethoxvDhenvnDterine 

A suspension of the compound of example 101 (10.46 g, 35 mmoles) in 
acetic anhydride (600 ml) and acetic acid (200 ml) was refluxed for 1 hour until 
a clear solution was formed By cooling down the reaction mixture in the 
5 refrigerator, the precipitate formed was filtered off, washed with ethyl acetate 
and diethyl ether, and then dned over P2O5 under vacuum, providing the title 
compound as a yellow powder (9.19 g, yield: 77 %). This compound was 
further characterized by the following spectra: 
- MS: m/z(%): 300 ([M+H]*, 100); 
10 - 'H-NMR (200 MHz, DMSO-d 6 ): 5 2.22 (3 H, s), 3.84 (3 H, s). 3.87 (3 H, 
s), 7.14 (1 H, d), 7.75 (2 H, m) and 9.51 (1 H, s) ppm. 

Example 103 - synthesis of 2^acetvlamin&4-(1.2.4-triazolvt^-6-f3.4-dimethoxv- 
phenvQpteridine 

15 To a solution of phosphorus oxychloride (1.68 ml, 18 mmoles) and 

1,2,4-tnazole (4.96 g, 72 mmoles) in dry pyridine (110 ml) was added the 
compound of example 102 (2.45 g, 7.18 mmoles). The suspension was stirred 
at room temperature for 4 hours. The precipitate was filtered off, washed with 
pyridine, toluene and diethyl ether. The resulting solid was dried over P2O5 

20 under vacuum, providing the title compound as a yellow powder (2 g, yield: 80 
%) which afforded the following mass spectrum 392 ([M+H]*, 100). 

Examples 104 and 105 - synthesis of 2-amino-4-mercaptoethvl-6-(3.4- 
dimethoxvphenvltoteridine and 2-aminc-4-mercaptoisopropvl-6-f3.4- 

25 dimethoxvphenvn ptendjng 

To a suspension of the compound of example 103 (0.25 mmole, 100 
mg) in dioxane (5 ml) was added 1 mmole of either ethanethiol (example 104) 
or isopropanethiol (example 105) and sodium (12 mg, 0.5 mmole). The 
suspension was stirred for 24 hours at room temperature. The solvent was 

30 concentrated in vacuo and the residue purified by flash chromatography 
(silica, using a CH3OH/CH2CI2 mixture (5:95) as an eluent), followed by 
purification by preparative TLC, providing the pure title compounds as yellow 



49 



powders with yields ranging from 20 to 30%. Both compounds were 
characterized by their mass spectrum as follows: 

- 2-amino4-mercaptoethyl-€-{3,4-dimethoxyphenyl) pteridine: 344 
([M+H]*, 100); 

5 - 2^mino-4-rnercaptoisopropyl-6-(3,4-dimethoxyphenyl) pteridine- 357 
([M+Hf, 100). 

Example 106 - synthesis of a mixture of 2.4<liamino-6-(PHnethoxvphenvl) 
pteridine and 2.4^iamino-7^p^ethoxvphenvl)pteridine 

10 2 f 4,5,64etra-aminopyrimidine (10 mmoles, 1.4 g) was dissolved in water (50 
ml) and the pH was adjusted to 9 with ammonium hydroxide. A solution of 4- 
methoxyphenylglyoxal (11 mmoles, 1.8 g) in ethanol (10 ml) was added 
dropwise and the solution was refluxed for 1 hour. The yellow precipitate 
formed was filtered off and washed with water, ethanol and diethyl ether. NMR 

IS analysis reveals the obtention of a mixture (1 2 g, 45 % yield) consisting of 87 
% of 2,4-diamino-7-(p-methoxyphenyl)pteridine and 13 % of 2,4-diamino-7-(p- 
methoxy-phenyl)pteridine. 1 H-NMR (500 MHz, TFA): 5 4.04 (3 H, s), 4.08 (3 H, 
s) t 7.15 (2 H, d) t 7.25 (2 H, d), 8.19 (2 H, d), 8.30 (2 H, d), 9.27 (1 H, s) and 
9 37 (1 H, s) ppm 

20 

Example 107 - synthesis of a mixture of 2^amino6-(p-methoxvphenvl)pterin 
and 2-amino-7-(xHnethoxyphenynpterin 

The mixture obtained in example 106 (1.2 g, 4.5 mmoles) was 
suspended in NaOH 1 N (80 ml) and refluxed till a solution was obtained The 
25 hot solution was treated with acetic acid till pH 5, then cooled down and the 
resulting precipitate was filtered off and washed with water, ethanol and 
diethyl ether, providing a mixture of 2-amino-6-(p-methoxyphenyl)pterin and 2- 
amino-7-(p-methoxyphenyl) pterin as a yellow powder (1 g, yield. 82 %). Mass 
spectrum: 270 ([M+Hf, 100). 

30 

Example 108 - synthesis of 2-acetylamino~6-{p-methoxvphenvl)pterin and 2- 
acetvlamino-7"fPHnethoxvphenvl)pterin 



50 

A suspension of the mixture obtained in example 107 (7.43 mmoles, 2 
g) was suspended in a mixture of acetic anhydride (50 ml) and acetic acid (50 
ml). The suspension was refluxed for 4 hours till a dear solution was obtained. 
Some insoluble material was filtered off and the solution was partly 
5 evaporated till precipation starts. Further precipitation was achieved overnight 
in the refrigerator. The resulting precipitate was filtered off and washed with 
ethyl acetate and diethyl ether, providing a mixture of 2-acetylamino-6-(p- 
methoxyphenyl)pterin and 2-acetylamino-7-(p-methoxyphenyl)pterin as a 
yellow powder (2.1 g, 91 % yield). Mass spectrum: 312 ([M+H]*, 100). 

10 

Example 109 - synthesis of 2-acetvlamino-4-f1.2.4-triazolvn-6-fp-methoxv- 
Phenvh pteridine and 2-acetvlaminc~4-n.2.4-triazolvlWp-methoxvDhenvn 
pteridine 

To a suspension of the mixture obtained in example 108 (1.5 g, 4 
15 mmoles) in dry pyridine (100 ml) was added 1 ,2,4-triazole (830 mg, 12 
mmoles) and 4-chlorophenyl phosphorodichloridate (1 ml, 6 mmoles). The 
suspension was stirred for 2 days at room temperature under nitrogen. The 
solvents were removed in vacuo. The solid matenal was suspended in 
dichloromethane and washed with 2 % HCI. Evaporation of the solvents 
20 provided a mixture of 2-acetylamirK>4-<1 ,2,4-triazolyl)-6-(p-methoxyphenyl) 
pteridine and 2-acetylamino-4-(1,2,4-triazolyl)-7-(p-methoxyphenyl)pteridine. 

Example 110 - synthesis of 2-amino-4HSODroooxv-7-fp-methoxvphenvn 
pteridine 

25 To a suspension of the mixture obtained in example 109 (180 mg, 0.50 

mmole) in isopropanol (8 ml) was added sodium (23 mg, 1 mmole). The 
suspension was stirred at room temperature overnight. The solvents were 
evaporated and the residue was purified by preparative TLC (silica, using a 
methanol/CH2CI 2 (7:93) mixture as the eluent). At this stage, both regio- 

30 isomers obtained were separated, thus providing the pure title compound as a 
yellow powder (yield- 45 %) which was further characterized by its mass 
spectrum: 312 ([M+Hf, 65), 270 flM+H-propene]*, 100). 



51 

Example 111 - synthesis of 2-amino-4HsoproDoxv-7-f3.4-dimethoxvphenvn 
pteridine 

The sequence of reactions described in examples 106 to 110 was 
followed, however starting from 3,4-dimethoxyphenylglyoxal instead of 4- 
5 methoxyphenylglyoxal in the first step. This provided 2-amino-4-isopropoxy-7- 
(3,4-dimethoxyphenyl) pteridine, a compound which was further characterized 
by its mass spectrum: 342 ([M+Hf. 55), 300 ([M+H-propenef, 75). 

Example 112 - synthesis of 2-amino^4-ethoxv-7-<3.4-dimethoxvphenvn 
10 ptendine 

The sequence of reactions described in examples 106 to 110 was 
followed, however starting from 3,4-dimethoxyphenylglyoxal instead of 4- 
methoxyphenylglyoxal in the first step, and from ethanol instead if isopropanol 
in the last step. This provided 2-amino-4-ethoxy-7-(3,4-dimethoxyphenyl) 
15 pteridine, a compound which was further characterized as follows" 

- MS: 328 ([M+HJ\100), 300 ([M+H-ethenef , 40); 

- 'H-NMR (500 MHz, DMSO-cfe): 6 1.44 (3 H, t), 3.86 (3 H, s), 3.88 (3 H, 
s), 4.54 (2 H, q), 7.13 (1 H. d), 7.16 (2 H, br s), 7.85 (1 H, d), 7.88 (1 H, 
dd) and 9.06 (1 H,s)ppm; 

20 - 13 C-NMR (125 MHz. DMSO-d 6 ): 6 14.25, 5567, 55.76, 63.06, 110.39, 
111.89, 121.13, 121.25, 128.24, 136.87, 149.28, 151.62, 155.82, 
156.72, 162 03 and 166.70 ppm. 

Example 113 - synthesis of 2^amino-4-methoxv-7-(3.4-dimethoxvphenvn 
25 pteridine 

The sequence of reactions described in examples 106 to 110 was 
followed, however starting from 3,4-dimethoxyphenylglyoxal instead of 4- 
methoxyphenylglyoxal in the first step, and from methanol instead if 
isopropanol in the last step. This provided 2-amino-4-ethoxy-7-(3,4- 
30 dimethoxyphenyl) pteridine, a compound which was further characterized by 
its mass spectrum: 314 ([M+Hr,100), 300 ([M+H-methanef, 20). 



EXAMPLE 1 14 - synthesis of 3.4-dimethoxvphenvlglvoxalmonoxime 



52 

Se0 2 (0.33 mole) was heated to 50°C in a mixture of dioxane (250 ml) and 
water (10 ml) After dissolution of Se0 2 , 3,4-dimethoxyacetophenone (0.3 
mole) was added and the mixture heated under reflux for 16 hours. The hot 
solution was filtered in order to remove selenium, the filtrate was evaporated, 
5 the oily residue dissolved in CHCI 3 (300 ml), then washed with saturated 
NaHC0 3 solution (100 ml) and water. The organic phase was dned over 
Na2S20 4 , filtered and evaporated. The yellow oil was distilled in vacuum, the 
resulting 3,4-dimethoxyphenylglyoxal was dissolved in methanol (50 ml) and 
water (200 ml), then acetonoxime (0.25 mole) was added and the pH adjusted 
10 to 4 by 2 N HCI. The solution was heated to 50°C for 2 hours, then cooled to 
0°C and the resulting crystals collected. After washing with cold water and 
drying in a vacuum desiccator, 3,4-dimethoxyphenylglyoxalmonoxime was 
obtained with a yield of 71 %, optionally recrystallized from CHCI 3 or acetone, 
and characterized by 'H-NMR (200 MHz, DMSO-cfe) showing peaks at 3.84 (3 
15 H, s), 7.06 (1 H, d), 7.51 (1 H, s), 7.75 (1 H, d), 8.10 (1 H, s) and 12.51 (1 H, 
s) ppm. 

Example 115 - alternative synthesis of 2-amino4-isoDroooxv-6-/3 4- 
dimethoxvphenyDpteridine 

20 To a suspension of the compound of example 59 (1.16 g, 6.34 mmole) in 
isopropanol (1.25 M HCI, 30 ml) was added the compound of example 114 
(6.34 mmole, 1 32 g). The reaction mixture was refluxed for 5 hours, then 
cooled down and the pH was adjusted to 9 by the addition of an aqueous 
concentrated solution of NH3 The precipitate was filtered off and further 

25 purified by flash chromatography over silica gel, using a isopropanol/C^Cb 
(1:99 to 3:97) mixture as the eluent, thus providing as a yellow powder 1.34 g 

of 2-amino-4-isopropoxy-6-(3,4-dimethoxyphenyl)pteridine, i.e. the compound 
of example 98 (yield: 62 %). 

30 Example 116 - synt hesis of 2.6-diamino^4-(1.2.3.6-tetrahvdropvridinvlV- 
pyrimidine 

To a suspension of 6-chtoro-2,4-diaminopyrimidine (6 g, 41 mmole) in toluene 
(50 ml) was added 1 ,2,3,6-tetrahydropyridine (8.23 ml, 91 mmole). The 



53 

resulting suspension was heated to reflux until a solution was obtained, then 
the solution was refluxed for 5 hours The reaction mixture was cooled down 
and water was added. The precipitate formed was filtered off and washed with 
toluene, providing 2,6-diamina4-(1,2 r 3,6-tetrahydropyridinyl)-pyrimidine as a 
5 white powder (7.4 g, yield 94 %). 

Example 117 - synthesis of 5-nitroso-2.6-diamino4-(1,2.3.6-tetrahvdro- 
pyridinvltovrimidine 

To a solution of the compound of example 1 16 (7.4 g, 38.7 mmole) in 
10 water (80 ml) and acetic acid (3.87 ml) was added a solution of sodium nitrite 
(2 94 g, 42.6 mmole) in water. The pink precipitate formed was filtered off and 
washed with water, providing 5-nitroso-2,6-diamino-4-(1,2,3,6-tetrahydro- 
pyridinyl)pyrimidine (7.83 g) with a yield of 92 %. 

15 Example 1 18 - synthesis of 2.5.6-triamino4-f1.2.3.6-tetrahvdroPvridinvn- 
pyrimidine 

To a suspension of the compound of example 117 (4 g, 18 mmole) in 
water (40 ml) was added sodium dithionite (7.9 g, 46 mmole). The suspension 
was heated to 90 °C until a solution was obtained. After cooling down, the 
20 2,5,6-tnamino-4-(1 ,2,3,6-tetrahydropyridinyl)pyrimidine precipitate formed was 
filtered off and used as such for further reaction. 

Example 119 - synthesis of 2-amino^4-(1.2.3.6-tetrahvdropvridinvl>-6-f3.4- 
dimethoxvphenyl)pteridine 

25 To a solution of the reaction product of example 118 (2.22 g, 10.8 mmole) in 
methanol (with 1 N HCI, 50 ml) was added the compound of example 114 
(2.26 g, 10.8 mmole). The solution was refluxed for 3 hours. The pH of the 
reaction was adjusted to 8 by the addition of an aqueous concentrated 
ammonia solution. The resulting precipitate was filtered off and further purified 

30 by flash chromatography over silica gel, using a CH 3 OH/CH 2 CI 2 mixture (3.97) 
as the eluent. This provided a yellow powder (2.56 g, yield 65 %) of 2-amino- 
4-(1,2,3,6-tetrahydropyridinyl)-6-(3,4-dimethoxyphenyl)pteridine which was 
characterized by the following spectra 



54 

- 'H-NMR (200 MHz, DMSO-d*) 5 3,84-3.88 (3 H, s), 4.45 (2 H, s), 4.75 
(2 H, s), 5.85-5.94 (1 H, m), 7.12 (2 H, d), 7.40 (2 H. s), 7.66 (1 H, s), 
7.72 (2 H, d) and 9.39 (1 H, s) ppm; 

- MS:365([M+Hf, 100). 

5 

Example 120 - TNF-g and IL-1 6 assays 

Peripheral blood mononuclear cells (herein referred as PBMC), in 
response to stimulation by lipopolysaccharide (LPS), a gram-negative 
bacterial endotoxin, produce various chemokines, in particular human TNF-a 
10 and IL-1 p. The inhibition of the activation of PBMC can be measured by the 
level of suppression of the production of TNF-a or IL-1 p by PBMC in 
response to stimulation by LPS 

Such inhibition measurement was performed as follows: PBMC were 
isolated from heparinized peripheral blood (Buffy coat) by density gradient 

15 centrifugation. LPS is then added to the PMBC suspension in complete 
medium (10 6 cells /ml) at a final concentration of 1 vgfml The pteridine 
derivative to be tested was added at different dilution levels, and the cells 
were incubated at 37°C for 72 hours. The supematants were collected, and 
TNF-a or IL-ip concentrations were measured with respectively an anti-TNF 

20 antibody or an anti-IL-1 p antibody in a sandwich ELISA (Duo Set ELISA 
human TNFa, commercially available from R&D Systems, United Kingdom). 
The colorimetric reading of the ELISA was measured by a Multiskan RC plate 
reader (commercially available from ThermoLabsystems, Finland) at 450 nm 
(reference wavelength 690 nm). Data analysis was performed with Ascent 

25 software 2.6. (also from ThermoLabsystems, Finland): a standard curve 
(recombinant human TNFa) was drawn and the amount (pg/ml) of each 
sample on the standard curve was determined. 

The % inhibition of human TNF-a production or human IL-1p was 
calculated using the formula : 

30 % inhibition = (pg/ml in sample - pg/ml min.) / (pg/ml max. - pg/ml min.) -1 
wherein: - min * pg/ml in culture medium without test compound, and 
- max.: pg/ml in culture medium + LPS without test compound. 



55 

Tables 1 and 2 hereinafter show the IC50 values (expressed in uM) of the 
tested compounds, being represented by the general formula (I), in these 
TNF-a and IL-1 assays. 



TABLE 1 



Example n° 


TNF-a 


IL-1 


24 


0.4 


5.5 


85 


6.7 


>40 


98 


3.5 


>40 


119 


0.5 


15 



5 

TABLE 2 



Example n° 


TNF-a 


Example n° 


TNF-a 


59 


8.0 


83 


6.6 


63 


4.0 


85 


6.7 


65 


9.1 


87 


7.5 


66 


4.5 


89 


9.1 


67 


10.0 


90 


6.6 


68 


10.0 


91 


6.2 


72 


8.5 


92 


10.0 


77 


8.1 


94 


7.6 


78 


8.5 


95 


6.3 


80 


68 


97 


9.1 


81 


10.0 


98 


35 



Example 121 - protection against lethal toxic shock. 

When a control group of 14 sham treated (saline injection) C3H mice 

10 are injected intraperitoneously with 100 pg LPS per mouse, all animals die 
within 1-3 days after injection. However when a group (16 animals) of the 
same C3H mice were treated for 2 days with the ptendine derivative of 
example 24 (intraperitoneous administration of 20 mg/kg/day; a first injection 
at the same time as the LPS injection, a second injection 24 hours later), all 

15 mice were significantly protected from acute shock related mortality: 



56 

- 14 animals permanently survived the challenge of a LPS lethal dose, 
and 

- 2 animals showed a significantly prolonged survival (5 days). 

Thus it can be concluded that treatment with a pteridine derivative of the 
5 invention provides an unexpectedly nearly complete protection against lethal 
toxic shock. 

Example 122 - prote ction aoainst a lethal dose of TNF-q 
A model of TNF-a induced shock in C57BL/6 male mice was performed as 

1 0 follows. Animals from the control group received an intravenous administration 
of a lethal dose of TNF-a (10 ug) in the tail. Animals from the test group 
received three intraperitoneous injections of a pteridine derivative (20 
mg/kg/day) respectively 48 hours, 24 hours and immediately before an 
intravenous injection of TNF-a (10 ug). 

15 Body temperature, a clinical sign of TNF-induced shock, was followed for 
48 hours in control mice and in mice receiving the pteridine derivative of 
example 24: the body temperature of control mice dropped significantly 
(28.2X) when compared to mice receiving the test compound (30.1°C). 
The survival rate (at least 50%) of mice that received the pteridine 

20 derivatives of the invention in addition to the TNF-a dose (10 ug) was quite 
substantial, as shown in table 3. 

TABLE 3 



Example n° 


Total number of 
animals 


Animals surviving 48 hours 
after TNF-a administration 


control 


9 


0 


24 


9 


7 


98 


6 


3 


119 


6 


3 



57 

This model therefore demonstrates that in vivo treatment with a 
pteridine derivative of this invention provides substantial and significant 
protection against a lethal dose of TNF-a. 

5 Example 123 - reduction o f tumor growth while inhibiting TNF-a toxicity 

A tumor model of melanoma (B16BL76) in C57BL6 (B6) mice was 
performed as follows. On day 1 , a group of 18 B6 mice were injected with 1 .5 
x 10 6 B16BU6 melanosarcoma cells. The group was further divided into the 
following sub-groups 3 days after the tumor cells injection: 
10 - a first control sub-group of 6 mice received vehicle (physiological 
solution) 3 times a week starting on day 3; 
- a second control sub-group of 5 mice received TNF-a (15 ug) on day 5 
and, for surviving animals, TNF-a was again administered 3 times a 
week for 2 weeks; 

15 - a test sub-group of 7 mice intraperitoneously received the pteridine 
denvative of example 24 at a dose of 20 mg/kg on each of days 3, 4 
and 5; the latter third injection on day 5 occurred 2 hours before a first 
TNF-a intravenous injection (15 ug). Then the test compound (20 
mg/kg/day) and TNFa (15 UQ/day) were administered each 
20 subcutaneously3timesaweekfor2weeks. 

Substantial protection against TNF-a toxicity was achieved by the pteridine 
derivative of the invention in tumor bearing mice: 5 out of 7 animals of the test 
sub-group survived, compared to none of the second control group (which all 
died during day 5). 

25 Furthermore, histological studies after two weeks treatment show that 
administration of the pteridine derivative of example 24 together with TNF-a 
leads to a significant reduction of tumor growth: the average tumor size (tumor 
size was measured as the largest diameter multiplied by the smallest 
diameter) in the test sub-group was 144 mm 2 , whereas the average tumor size 

30 in the first control group was 439 mm 2 These data clearly demonstrate that 
the pteridine derivative of example 24 effectively protects mice against the 
toxicity of TNF-a while preserving its anti-tumor effects. 



58 



CLAIMS 



1. Use of a pteridine derivative for the manufacture of a medicament for the 
prevention or treatment of a disorder in a mammal, the said disorder being 
selected from the group consisting of: 

- septic or endotoxic shock, 

- TNF-a- mediated diseases, 

- pathologies and conditions associated with and/or induced by abnormal 
levels of TNF-a occurring in a systemic, localized or particular tissue type or 
location in the body of the mammal, 

- toxic effects of TNF-a and/or anti-cancer chemotherapeutic agents, 

- injuries after irradiation of a tissue of the mammal by radio-elements, and 

- cachexia, 

the said pteridine derivative having the general formula (I): 



wherein X represents an oxygen atom or a group with the formula S(0) m wherein m 
is an integer from 0 to 2, or a group with the formula NZ and wherein: 

- Ri is a group selected from the group consisting of C1-7 alkyl, C2.7 alkenyl, 
C2-7 alkynyl, C3-10 cycloalkyl, C3.10 cycloalkenyl, aryl, alkylaryl, arylalkyl, 
heterocyclic, heterocyclic-substituted alkyl and alkyl-substituted 
heterocyclic, each of said groups being optionally substituted with one or 
more substituents selected from the group consisting of halogen, Ci^ 
alkyl, C1.4 alkoxy, C 2 . 7 alkenyl, C2-7 alkynyl, halo C1.4 alkyl. C3.10 
cycloalkoxy, aryloxy, arylalkyloxy, oxyheterocyclic, heterocyclic-substituted 
alkyloxy, thio C,. 7 alkyl, thio C3-10 cycloalkyl, thioaryl, thioheterocyclic, 
arylalkylthio, heterocyclic-substituted alkylthio, formyl, hydroxyl, suKhydryl, 
nitro, hydroxylamino, mercaptoamino, cyano, carboxylic acid or esters or 
thioesters or amides or thioamides or halides or anhydrides thereof, 
thiocarboxylic acid or esters or thioesters or amides or thioamides or 




58 



59 



halides or anhydrides thereof, carbamoyl, thiocarbamoyl, ureido, thio- 
ureido, amino, cycloalkylamino, alkenylamino, cycloalkenylamino, 
alkynylamino, arylamino, arylalkyl-amino, hydroxylalkylamino, 
mercaptoalkyl-amino, heterocyclic amino, hydrazino, alkylhydrazino and 
phenyl-bydrazino; or R 1 is a carboxyalkyl, carboxyaryl, thiocarboxyaryl or 
thiocarboxyalkyl group; 

Z is a group independently defined as Ri or Z is hydrogen or the group NZ 
together with Ri is either hydroxylamino or an optionally substituted 
heterocyclic group containing at least one nitrogen atom; 
R2 is selected from the group consisting of amino; acylamino; 
thioacylamino, carbamoyl; thiocarbamoyl, ureido; thioureido, sulfon-amido; 
hydroxylamino; alkoxyamino; thioalkylamino; mercaptoamino, hydrazino; 
alkylhydrazino; phenylhydrazino; optionally substituted heterocyclic 
radicals, C1-7 alkylamino, arylamino; arylalkylamino; cycloalkylamino; 
alkenylamino; cycloalkenylamino; heterocyclic amino; hydroxyalkylamino; 
mercaptoalkylamino; C1-7 alkoxy; C3.10 cycloalkoxy; thio C1.7 alkyl; 
arylsulfoxide; arylsulfone; heterocyclic sulfoxide; heterocyclic sulfone; thio 
C3-10 cycloalkyl; aryloxy; arylthio; arylalkyloxy; arylalkylthio; oxyheterocyclic 
and thioheterocyclic radicals, 

R4 is an atom or a group selected from the group consisting of hydrogen; 
halogen, C1-7 alkyl; C2-7 alkenyl; C2-7 alkynyl; halo C1.7 alkyl; carboxy C1-7 
alkyl; acetoxy C1-7 alkyl; carboxyaryl; C1-7 alkoxy; C3-10 cycloalkoxy; 
aryloxy; arylalkyloxy; oxyheterocyclic; heterocyclic-substituted alkyloxy, 
thio C1-7 alkyl, thio C3.10 cycloalkyl; thioaryl; thioheterocyclic; arylalkylthio; 
heterocyclic-substituted alkylthio; amino; hydroxylamino; mercapto-amino; 
acylamino, thioacylamino, alkoxyamino; thioalkylamino; acetal; thioacetal; 
carboxylic acid; carboxylic acid esters, thioesters, halides, anhydrides, 
amides and thioamides, thiocarboxylic acid; thiocarboxylic acid esters, 
thioesters, halides, anhydrides, amides and thioamides; hydroxyl, 
sulfhydryl; nitro; cyano; carbamoyl; thiocarbamoyl, ureido; thio-ureido; 
alkylamino; cycloalkylamino; alkenylamino; cycloalkenylamino; alkynyl- 



59 



60 



amino; arylamino; arylalkylamino; hydroxyalkylamino; mercapto- 
alkylamino; heterocyclic amino; heterocyclic-substituted alkylamino, 
oximino; alkyloximino; hydrazino; alkylhydrazino; phenylhydrazino; 
cysteinyl add, esters, thioesters, halides, anhydrides, amides and 
thioamides thereof; aryl groups optionally substituted with one or more 
substituents selected from the group consisting of halogen, C1.7 alkyl, C1-7 
alkoxy, C2-7 alkenyl, C 2 -? alkynyl, halo C1.7 alkyl, nitro, hydroxyl, sulfhydryl, 
amino, C3.10 cycloalkoxy, aryloxy, arylalkyloxy, oxyhetero-cyclic, 
heterocyclic-substituted alkyloxy, thio C1-7 alkyl, thio C^o cycloalkyl. 
thioaryl, thioheterocyclic, arylalkylthio, heterocyclic-substituted alkylthio, 
formyl, carbamoyl, thiocarbamoyl, ureido, thio-ureido, sulfonamido, 
hydroxylamino, alkoxyamino, mercaptoamino, thioalkylamino, acylamino, 
thioacylamino, cyano, carboxylic acid or esters or thioesters or halides or 
anhydrides or amides thereof, thiocarboxylic acid or esters or thioesters or 
halides or anhydrides or amides thereof, alkylamino, cycloalkylamino, 
alkenylamino, cycloalkenylamino, alkynylamino, arylamino, arylalkylamino, 
hydroxyalkylamino, mercaptoalkylamino, heterocyclic amino, hydrazino, 
alkyl-hydrazino and phenylhydrazino; optionally substituted heterocyclic 
radicals; aromatic or heterocyclic substituents substituted with an aliphatic 
spacer between the pteridine ring and the aromatic or heterocyclic 
substituent, whereby said aliphatic spacer is a branched or straight, 
saturated or unsaturated aliphatic chain of 1 to 4 carbon atoms which may 
contain one or more functions, atoms or radicals selected from the group 
consisting of carbonyl (oxo), thiocarbonyl, alcohol (hydroxyl), thiol, ether, 
thio-ether, acetal, thio-acetal, amino, imino, oximino, alkyloximino, amino- 
acid, cyano, acylamino, thioacylamino, carbamoyl, thiocarbamoyl, ureido, 
thio-ureido, carboxylic aad or ester or thioester or halide or anhydride or 
amide, thiocarboxylic acid or ester or thioester or halide or anhydride or 
amide, nitro, thio Ct-7 alkyl, thio C3.10 cycloalkyl, hydroxylamino, 
mercaptoamino, alkylamino, cycloalkylamino, alkenylamino, cycloalkenyl- 
amino, alkynylamino, arylamino, arylalkylamino, hydroxyalkylamino, 



60 



61 



mercaptoalkylamino, heterocyclic amino, hydrazino, alkythydrazino, 
phenylhydrazino, sulfonyl, sulfinyl, sulfbnamido and halogen; branched or 
straight, saturated or unsaturated aliphatic chains of 1 to 7 carbon atoms 
optionally containing one or more functions selected from the group 
consisting of carbonyl (oxo), thiocarbonyl, alcohol (hydroxyl), thiol, ether, 
thio-ether, acetal, thio-acetal. amino, imino, oximino, alkykoximino, amino- 
acid, cyano, acylamino; thioacylamino, carbamoyl, thiocarbamoyl, ureido, 
thio-ureido, carboxylic acid ester or halide or anhydride or amide, 
thiocarboxylic acid or ester or thioester or halide or anhydride or amide, 
nitro, thio Ci- 7 alkyl, thio C3-10 cycloalkyl, hydroxylamino, mercapto-amino, 
alkylamino, cycloalkylamino, alkenylamino, cycloalkenylamino, alkynyl- 
amino, arylamino, arylalkylamino, hydroxyalkylamino, mercaptoalkylamino, 
heterocyclic amino, hydrazine alkylhydrazino, phenylhydrazino, sulfonyl, 
sulfinyl, sulfonamido and halogen; and 

- R3 is an atom or a group defined as FU or R 3 together with R4 forms a 
homocyclic or heterocyclic radical; 

and/or a pharmaceutical^ acceptable addition salt thereof and/or a 

stereoisomer thereof and/or a mono- or a di-/V-oxide thereof and/or a solvate 

and/or a dihydro- or tetrahydropteridme derivative thereof. 

2. Use according to claim 1, wherein Ri is selected from the group consisting of 
methyl, ethyl, isopropyl and pentyl 

3. Use according to claim 1, wherein R 3 is 3-thienyl, 2-thienyl or a phenyl group 
with one or more substituents. 

4. Use according to claim 1, wherein R3 is a phenyl group with one or more 
substituents each independently selected from the group consisting of fluoro, 
methoxy, ethoxy, trifluoromethyl, dimethylamino, chloro, cyano, methyl, ethyl, 
carboxymethyl, methylthio, dimethylcarboxamido, diethylcarboxamido and 
methylcarboxylate. 



61 



62 

Use according to claim 1 , wherein: 

- XisNZ, 

- Z is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl 
and benzyl, and 

- Ri is selected from the group consisting of methyl, ethyl, n-propyl and 
benzyl. 

Use according to claim 1 , wherein X is NZ and wherein the group NZ together 
with Ri is selected from the group consisting of tetrahydropyridinyl, 
hydroxylamino, morpholinyl, piperidinyl, piperazinyl, 1,2,4-triazolyl and N- 
methylpiperazinyl. 

Use according to claim 1, wherein the pteridine derivative is a compound 
selected from the group consisting of. 

- 2-amino-4-ethoxypteridine 

- 2-amino-4-ethoxy-6-chlcro-pteridine 

- 2-amino-4-ethoxy-€-(4-methoxyphenyl)-pteridine 

- 2-amino-4-ethoxy-6-(2-methoxyphenyl)-pteridine 

- 2-amino-4-ethoxy-6-<3-methoxyphenyl)-pteridine 

- 2-amino-4-ethoxy-€-{3,4-difluorophenyl)-pteridine 

- 2-amino-4-ethoxy-6-(4-dimethylaminophenyl)-pteridine 

- 2-amino-4-ethoxy-6-(4-trifluoromethylphenyl)-pteridine 

- 2-amino4-ethoxy-6-(2-thienyl)-pteridine 

- 2-amino-4-ethoxy-6-(3-thienyl)-pteridine 

- 2-amino-4-ethoxy-6-{3,4-dichlorophenyl)-pteridine 
2-amino-4-ethoxy-6-<4-cyanophenyl)-pteridine 
2-amino-4-ethoxy-6-(4-ethoxyphenyl)-pteridine 
2-amino-4-ethoxy-6-(4-fluorophenyl)-pteridine 
2-amino-4-ethoxy-6-(4-ethylphenyl)-pteridine 
2-amino-4-ethoxy-6-(4-acetylphenyl)-pteridine 
2-aminc-4^oxy^3^uoro-4-methylphenyl)-pteridine 



62 



63 



2-amino-4-ethoxy-6-{4-methylthiophenyl)-pteridine 

2-amino-4-ethoxy-6-(4-N,N-dimethylbenzamido)-pter1dine 

2-amino-4-isopropoxypteridine 

2-amino4-isopropoxy-e<*)loropteridine 

2^mino4HSopropoxy^3HTiethyl-4-methoxyphenyl)-pteridine 

2-amino^sopropoxy^3,4-dmethylphenyl)-pteridine 

2^ino^sopropoxy^3^lor(M-trrfluoromethylphenyl)-pteridine 

2-amino^sopropoxy-6-<3-chlorol-4-fluorophenyl)-pteridine 

2^mino^sopropoxy^4-N,N-diethylbenzamido)-pteridine 

2-amino-^sopropoxy-6-(4-trifIuoromethylphenylH)teridine 

2-amino-4-isopropoxy-6-(3,4-difluorophenyl)-pteridine 

2-amino-4Hsopropoxy-6-(4-methoxyphenyl)-ptendine 

2-ammo-4-isopropoxy-6-(4-ethoxyphenyl)-pteridine 

2-amino^sopropoxy^4-N,N^imethylbenzamjdo)-pteridine 

2-amino-4-isopropoxy-6-{3-thienyl)-ptendine 

2^mino-4-isopropoxy-6-(4-cyanophenyl)-pteridine 

2-amino-4-isopropoxy-6-(4-benzoic add methyl ester)-pteridine 

2-amino-4-isopropoxy-6-(4-acetylphenyl)-pteridine 

2^mino^HSopropoxy^3,4^imethoxyphenyl)-pteridine 

2^mino^thylthio^3,4^imethoxyphenyl)-pteridine 

2^mincM^sopropylthio-6-<3,4-dimethoxyphenyl)-pteridine 

2^mincMHDentoxy-6-styiylpterjdine, 

2^min<M-n-pentoxy^1,2<libromo-2^ 

2^mino-4-melhoxy-6-styryl-7-methoxypteridine, 

2,4-diamino^-phenyl-7-methylpteridine, 

2-amino-4-dimethylamino-6-phenylpteridine, 

2^mino-4-dimethylarnino-6-{4-tolyl)pteridine 1 

2-amino-4-dimethylamino-6-(4-methoxyphenyl)pteridine, 

2-amino-4-diethylamino-6-phenylpteridine, 

2-amincH4-diethylamino^-(4-chlorophenyl)pteridine 1 

2^mino-4^1iethylamino-€-{4-methoxyphenyl)pteridine, 



64 



2^mino^iethylamino^3,4<limethoxyphenyl)pteridine l 

2-amino-4-dibenzylamino-6i3henyl pteridine, 

2-amino-4^ibenzylamino-^4-chlorophenyl)pteridine l 

2^mino-4^ibenzylamino^4HTiethoxyphenyl)pteridine 

2^mino-4^ibenzylaminch6^3,4^ 

2^mino-4-dipropylamino-6-phenylpteridine, 

2^mino-4^ipropylamino^4^lorophenyl)pteridine, 

2-amino-4^ipropylamino^4^ethoxyphenyl)pteridine, 

2^mino-4^ipropylamino^3 t 4^imethoxyphenyl)pteridine, 

2^mincHl^oipholino-6-phenylpteridine > 

2^mino-4^oipholino^^4-chlorophenyl)pteridine t 

2^mino-4^orpholino^4-methoxyphenyl)pteridine, 

2^mino-4^rpholino^3,4^imemo 

2-amino-4^iperidino-6-phenylpteridine, 

2-amino-4^iperidirK)^4-chlorophenyl) pteridine, 

2-amino-4^iperidino^4-methoxyphenyl)pteridine, 

2^mino^^iperidino^3,4Kiimethoxyphenyl)pteridine l 

2-amino-4-N-methylpiperazino-€-phenylptendine, 

2^mino^N^ethylpiperazino^4^l^^ 

2-amino^N-methylpiperazino^ 

2^mino-4^ethylpiperazino^3,4<limetnoxyphen 

2-amirK>-4^yrrolidino^4HTiethoxyphenyl)pterid 

2^mincM-piperazinc>-6-phenyl^eridine l 

2-amirw>-4^iperazirK>^4Kiilorophen^ 

2^mino-4i3ip€razirK>^4^ethoxyphenyl)pteridi 

2^mino-4^iperazirK>^3 l 4Klimethoxyphenyl)pteridine i 

2-amino^ibenzylamino^3,4 t 5-^ 

2^mincMHrnorpholino^(3 l 4 f 5-trimethoxyphenyl)pteridine ( 

2-amino^3-adamantylamino)^3 t 4 l 5-trimethoxyphenyO 

2-amino^3^amantylamino)-6-naphtylpteridine t 

2^mino^4^amantylamino)^3 l 4,5-trimethoxyphenyl)pterkjine i 



65 

- 2-amino-4-(4-adamantylamino)-6-naphtylpteridine > 

- 2^mino^^orpholino^3,4^ormylidene-3,4^ihydroxyphenyl)pteridine, 

- 2^mino^imethylamino^^3Aformylidene-3,4-dihydroxyphenyl) 
pteridine, 

- 2^mino-4-pyrrolidino-6-(3 1 4,dimethoxyphenyl)pteridine > 

- 2^mirx>^imethylamino-6-(3,4-dimethoxyphenyl)pteridine, 

- 2-amino-4-dimethylamino-6-methylptendine, 

- 2-amino-4-ethoxy-6-phenylpteridine, 

- 2^mino-4-propylamino^-phenylpteridine 1 

- 2^m«no-4^opylamino-6-{3,4<limethoxyphenyl)pteridine, 

- 2-acetamido-4-hydraxy-6-(3 1 4-dimethoxyphenyl)pteridine, 

- 2-acetamido-4Hsopropoxy-6-(3,4-dimethoxyphenyl)pteridine 1 

- 2-amino-4-ethoxy-6-(3,4-dimethoxyphenyl)pteridine 1 and 

- 2-amino-4-<1 ,2,3,6-tetrahydropyridinyl)-6-(3,4-dimethoxyphenyl)pteridine. 

8. Use according to claim 1, wherein the said disorder is septick shock and the 
medicament is for the treatment of a mammal with a serum level of 
interleukin-6 above 1 ,000 pg/ml at start of treatment. 

9. Use according to claim 1, wherein the said TNF-a-mediated disease is 
selected from the group consisting of neurodegenerative diseases, 
myelodysplastic syndromes and alcohol-induced hepatitis. 

10. Use according to claim 1, wherein the abnormal levels of TNF-a are levels 
exceeding by at least 10 % and at most 500% the TNF-a level present in a 
normal healthy subject. 



65 



o 

INVESTOR IN PEOPLE 

Darren Handley 
13 January 2004 

Patents Act 1977 : Search Report under Section 17 



Documents considered to be relevant: 



Category 


Relevant 
to claims 


Identity of document and passage or figure of particular relevance 


X 


1-3, 5-10 


DE 19944767 A 


(VASOPHARM) - see page 1, lines 17-50; page 
7, lines 15-25; and page 9, line 28-page 11, line 
60 


X 


MO 


JP 2003238409 A 


(SHIMICK) - see WPI abstract AN-2003- 
820621 [77] 


X 


1,8-10 


JP 6192100 A 


(ASHAHI) - see WPI abstract AN-1994-260430 
[32] and PAJ and paragraphs [0005]-[0007] 


X 


1.8-10 


US 5902810 A 


(PFLEIDERER) - see claims 1, 8-9; column 7, 
line 6-column 8, line 60 


A 




WO 00/39129 A 


(LEUVEN) - see claims 1 & 6 



Categories: 



X 


Document indicating lack of novelty or inventive step 


A 


Document indicating technological background and/or state of the art 


Y 


Document indicating lack of inventive step if combined 
with one or more other documents of same category. 


P 


Document published on or after the declared priority date but before the 
filing date of this invention. 


& 


Member of the same patent family 


E 


Patent document published on or after, but with priority date earlier 
than, the filing date of this application 



Field of Search: 

Search of GB, EP, WO & US patent documents classified in the following areas of fee UKC*: 



Worldwide search of patent documents classified in the following areas of the IPC 7 : 

A61K; A61P 

The following online and other databases have been used in the preparation of this search report: 
WPI, EPODOC, JAPIO, CAS-ONLINE 




Application No: 
Claims searched: 



Hit- *d 



♦~ s Patent 
X Office 



OS 



GB 0321384.0 
1-10 



Examiner: 
Date of search: 



An Executive Agency of the Department of Trade and Industry